CN116745288A - Compounds and uses thereof - Google Patents

Compounds and uses thereof Download PDF

Info

Publication number
CN116745288A
CN116745288A CN202180089830.4A CN202180089830A CN116745288A CN 116745288 A CN116745288 A CN 116745288A CN 202180089830 A CN202180089830 A CN 202180089830A CN 116745288 A CN116745288 A CN 116745288A
Authority
CN
China
Prior art keywords
cancer
compound
optionally substituted
pharmaceutically acceptable
acceptable salt
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202180089830.4A
Other languages
Chinese (zh)
Inventor
K·J·威尔逊
S·内格雷蒂
S·E·R·席勒
R·G·瓦斯瓦尼
D·S·黄
J·H·福格特
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Foghorn Treatment Co
Original Assignee
Foghorn Treatment Co
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Foghorn Treatment Co filed Critical Foghorn Treatment Co
Publication of CN116745288A publication Critical patent/CN116745288A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Abstract

The present disclosure features compounds for use in the treatment of BAF complex-related disorders.

Description

Compounds and uses thereof
Background
The present invention relates to compounds useful for modulating BRG1 or BRM-associated factor (BAF) complexes. In particular, the present invention relates to compounds useful for the treatment of disorders associated with BAF complex function.
Chromatin regulation is essential for gene expression, and ATP-dependent chromatin remodeling is the mechanism by which such gene expression occurs. Human switch/sucrose non-fermentive (SWI/SNF) chromatin remodeling complexes, also known as BAF complexes, have two SWI 2-like atpases known as BRG1 (Brahma-related gene-1) and BRM (Brahma). The transcriptional activator BRG1, also known as ATP-dependent chromatin remodeling factor SMARCA4, is encoded by the SMARCA4 gene on chromosome 19. BRG1 is overexpressed in some cancer tumors and is required for proliferation of cancer cells. BRM, also known as the possible global transcriptional activator SNF2L2 and/or ATP-dependent chromatin remodeling factor SMARCA2, is encoded by the SMARCA2 gene on chromosome 9 and has been shown to be essential for tumor cell growth in cells characterized by a loss of BRG1 function mutation. Inactivation of BRG and/or BRM results in downstream effects in cells, including cell cycle arrest and tumor suppression.
SUMMARY
The invention features compounds useful for modulating BAF complexes. In some embodiments, the compounds are useful for treating disorders associated with alterations to BAF complexes, such as disorders associated with alterations to one or both of BRG1 and BRM proteins. The compounds of the invention, alone or in combination with other pharmaceutically active agents, may be useful in the treatment of such disorders.
In one aspect, the invention provides a compound having the structure:
wherein the method comprises the steps of
m is 0, 1, 2 or 3;
n is 0, 1, 2, 3 or 4;
X 1 is-S-, -SO 2 -or-S (O) (NH) -;
X 2 is N or CR 8
R 1 For hydrogen or optionally takingSubstituted C 1 -C 6 An alkyl group;
each R 2 And each R 3 Independently hydrogen, optionally substituted C 1 -C 6 Alkyl or optionally substituted C 1 -C 6 A heteroalkyl group;
L 1 is an optionally substituted 9-to 10-membered bicyclic heterocyclyl or an optionally substituted 9-to 10-membered bicyclic heteroaryl;
L 2 absent, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted C 6 -C 10 Aryl, optionally substituted 5-to 14-membered heteroaryl, or optionally substituted 4-to 14-membered heterocyclyl;
R 4 is hydrogen, halogen, optionally substituted C 1 -C 6 Alkyl or optionally substituted C 3 -C 10 Cycloalkyl;
R 5 is optionally substituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl or optionally substituted amino, and R 6 Is hydrogen, halogen, cyano, optionally substituted C 1 -C 6 Alkyl, optionally substituted C 2 -C 6 Alkenyl or optionally substituted C 3 -C 10 Cycloalkyl; or R is 5 And R is 6 And together with the attached atoms, are combined into an optionally substituted 5-to 8-membered heterocyclyl;
each R 7 Independently optionally substituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl, halogen, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted C 3 -C 10 Cycloalkyl C 1 -C 6 Alkyl, optionally substituted 5-to 14-membered heteroaryl, optionally substituted 4-to 14-membered heterocyclyl, -N (R) 7A ) 2 OR-OR 7A Wherein each R is 7A Independently H, optionally substituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted C 6 -C 10 Aryl, optionally substituted 5-to 10-membered heteroaryl or optionally substituted 4-to 14-membered heterocyclyl, or two geminal R 7A The groups are combined with the attached atoms to form an optionally substituted 5-to 10-membered heteroaryl or an optionally substituted 4-to 10-membered heterocyclyl; or two twin R 7 The groups are combined with the attached atoms to form carbonyl groups;
R 8 is hydrogen, halogen, optionally substituted C 1 -C 6 Alkyl or optionally substituted C 3 -C 10 Cycloalkyl; and is also provided with
R 9 Hydrogen or halogen;
or a pharmaceutically acceptable salt thereof.
In some embodiments, the variables for the compounds of formula I are as follows:
m is 0, 1, 2 or 3;
n is 0, 1, 2, 3 or 4;
X 1 is S, SO 2 Or S (O) (NH);
X 2 is N or CR 8
R 1 Is hydrogen or optionally substituted C 1 -C 6 An alkyl group;
each R 2 And each R 3 Independently hydrogen, optionally substituted C 1 -C 6 Alkyl or optionally substituted C 1 -C 6 A heteroalkyl group;
L 1 is an optionally substituted 9-to 10-membered bicyclic heterocyclyl or an optionally substituted 9-to 10-membered bicyclic heteroaryl;
L 2 absent, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted C 6 -C 10 Aryl, optionally substituted 5-to 10-membered heteroaryl or optionally substituted 4-to 10-membered heterocyclyl;
R 4 is hydrogen, halogen, optionally substituted C 1 -C 6 Alkyl or optionally substituted C 3 -C 10 Cycloalkyl;
R 5 is optionally substituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl or optionally substituted amino, and R 6 Is hydrogen, halogen, cyano, optionally substituted C 1 -C 6 Alkyl or optionally substituted C 3 -C 10 Cycloalkyl; or R is 5 And R is 6 And together with the attached atoms, are combined into an optionally substituted 5-to 8-membered heterocyclyl;
each R 7 Independently optionally substituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl, halogen, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted 5-to 10-membered heteroaryl, optionally substituted 4-to 10-membered heterocyclyl, -N (R) 7A ) 2 OR-OR 7A Wherein each R is 7A Independently H, optionally substituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted C 6 -C 10 Aryl, optionally substituted 5-to 10-membered heteroaryl or optionally substituted 4-to 10-membered heterocyclyl, or two geminal R 7A The groups are combined with the attached atoms to form an optionally substituted 5-to 10-membered heteroaryl or an optionally substituted 4-to 10-membered heterocyclyl;
R 8 is hydrogen, halogen, optionally substituted C 1 -C 6 Alkyl or optionally substituted C 3 -C 10 Cycloalkyl; and is also provided with
R 9 Is hydrogen;
or a pharmaceutically acceptable salt thereof.
In some embodiments, L 2 Absent, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted C 6 -C 10 Aryl, optionally substituted 5-to 10-membered heteroaryl, or optionally substituted 4-to 10-membered heterocyclyl.
In some embodiments, each R 7 Independently optionally substituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl, halogen, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted 5-to 10-membered heteroaryl, optionally substituted 4-to 10-membered heterocyclyl, -N (R) 7A ) 2 OR-OR 7A Wherein each R is 7A Independently H, optionally takenSubstituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted C 6 -C 10 Aryl, optionally substituted 5-to 10-membered heteroaryl or optionally substituted 4-to 10-membered heterocyclyl, or two geminal R 7A The groups are combined with the attached atoms to form an optionally substituted 5-to 10-membered heteroaryl or an optionally substituted 4-to 10-membered heterocyclyl.
In some embodiments, R 5 And R is 6 And are combined with the attached atoms to form an optionally substituted 5-to 8-membered heterocyclic group. In some embodiments, R 5 And R is 6 And are combined with the attached atoms to form an optionally substituted 7-membered heterocyclic group.
In some embodiments, R 5 Is optionally substituted C 1 -C 6 An alkyl group. In some embodiments, R 5 Is an optionally substituted amino group. In some embodiments, R 6 Is optionally substituted C 1 -C 6 An alkyl group. In some embodiments, R 6 Is halogen.
In some embodiments, X 1 Is SO 2 . In some embodiments, X 2 Is CR (CR) 8
In some embodiments of the present invention, in some embodiments,
a group of the structure->Wherein the method comprises the steps of
Z is CH 2 CO or C (R) X2 ) 2
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups;
each R X2 Independently H or optionally substituted C 1 -C 6 An alkyl group; and is also provided with
p is 0, 1, 2, 3 or 4.
In some embodiments of the present invention, in some embodiments,a group of the structure->Wherein the method comprises the steps of
Z is CH 2 CO or C (R) X2 ) 2
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups;
each R X2 Independently H or optionally substituted C 1 -C 6 An alkyl group; and is also provided with
p is 0, 1, 2, 3 or 4.
In some embodiments of the present invention, in some embodiments,a group of the structure->Wherein the method comprises the steps of
Z is CH 2 CO or C (R) X2 ) 2
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups;
each R X2 Independently hydrogen or optionally substituted C 1 -C 6 An alkyl group;
p is 0, 1, 2, 3 or 4; and is also provided with
q is 0 or 1.
In some embodiments of the present invention, in some embodiments,a group of the structure->Wherein the method comprises the steps of
Z is CH 2 CO or C (R) X2 ) 2
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups;
each R X2 Independently hydrogen or optionally substituted C 1 -C 6 An alkyl group; and is also provided with
p is 0, 1, 2, 3 or 4.
In some embodiments of the present invention, in some embodiments,a group of the structure->Wherein the method comprises the steps of
Is a single bond or a double bond;
each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups;
R X2 is hydrogen or optionally substituted C 1 -C 6 An alkyl group; and is also provided with
p is 0, 1, 2, 3 or 4.
In some embodiments of the present invention, in some embodiments,a group of the structure- >Wherein the method comprises the steps of
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl orHalogen, or two twin R X1 The groups are combined with the attached atoms to form carbonyl groups;
R X2 is hydrogen or optionally substituted C 1 -C 6 An alkyl group; and is also provided with
p is 0, 1, 2, 3 or 4.
In some embodiments, R 8 Is hydrogen.
In some embodiments, R 8 Is halogen.
In some embodiments, R 8 Is optionally substituted C 3 -C 8 Cycloalkyl groups.
In some embodiments, X 2 Is N.
In some embodiments of the present invention, in some embodiments,a group of the structure->Wherein the method comprises the steps of
Z is CH 2 CO or C (R) X2 ) 2
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups;
each R X2 Is independently hydrogen or optionally substituted C 1 -C 6 An alkyl group; and is also provided with
p is 0, 1, 2, 3 or 4.
In some embodiments of the present invention, in some embodiments,a group of the structure->Wherein the method comprises the steps of
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 With groups bound theretoTogether atoms are combined into carbonyl or C 3 -C 8 Cycloalkyl ring, or two ortho-positions R X1 The radicals being further combined with the atoms to which they are attached to form C 3 -C 8 A cycloalkyl ring;
p is 0, 1, 2, 3 or 4; and is also provided with
q is 0, 1 or 2.
In some embodiments of the present invention, in some embodiments,a group of the structure- >Wherein the method comprises the steps of
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups; and p is 0, 1, 2, 3 or 4.
In some embodiments, at least one R X1 Is optionally substituted C 1 -C 6 An alkyl group.
In some embodiments, -L 2 -(R 7 ) n Is a group of the structure:
in some embodiments, at least one R X1 Is halogen.
In some embodiments, at least two twins R X1 The groups combine with the attached atoms to form carbonyl groups.
In some embodiments, L 1 Is an optionally substituted 9-to 10-membered bicyclic heteroaryl.
In some embodiments, L 1 Is that
Wherein the method comprises the steps of
X 3 、X 4 、X 5 、X 6 、X 7 And X 8 Each independently is N or CR L1
Each R L1 Independently H, halogen, optionally substituted C 1 -C 6 An alkyl group;
A 1 bonding to- (C (R) 2 )(R 3 )) m -; and is also provided with
A 2 Bonded to L 2
In some embodiments, L 1 Is that
In some embodiments, L 1 Is that
In some embodiments, L 1 Is that
In some embodiments, L 1 Is that
In some embodiments, L 1 Is that
In some embodiments, L 1 Is thatWherein A is 1 Bonding to- (C (R) 2 )(R 3 )) m -; and A is 2 Bonded to L 2
In some embodiments, L 2 Is an optionally substituted 5-to 10-membered heteroaryl.
In some embodiments, -L 2 -(R 7 ) n Is a group of the structure:
In some embodiments, -L 2 -(R 7 ) n Is a group of the structure:
in some embodiments, -L 2 -(R 7 ) n Is a group of the structure:
in some embodiments, -L 2 -(R 7 ) n Is a group of the structure:
in some embodiments, -L 2 -(R 7 ) n Is a group of the structure:
in some embodiments, -L 2 -(R 7 ) n Is a group of the structure:
in some embodiments, -L 2 -(R 7 ) n Is a group of the structure:
in some embodiments, -L 2 -(R 7 ) n Is a group of the structure:
in some embodiments, -L 2 -(R 7 ) n Is a group of the structure:
in some embodiments, -L 2 -(R 7 ) n Is a group of the structure:
in some embodiments, L 2 Is optionally substituted C 6 -C 10 Aryl groups.
In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments, n is 3.
In some embodiments, R 7 Is optionally substituted C 1 -C 6 An alkyl group. In some embodiments, R 7 Is optionally substituted C 1 -C 6 A heteroalkyl group. In some embodiments, R 7 Is an optionally substituted 4-to 14-membered heterocyclyl. In some embodiments, R 7 Is optionally substituted azetidinyl or optionally substituted morpholinyl. In some embodiments, R 7 Is optionally substituted C 3 -C 10 Cycloalkyl groups. In some embodiments, R 7 Is optionally substituted cyclopropyl or optionally substituted cyclobutyl. In some embodiments, R 7 is-N (R) 7A ) 2 . In some embodiments, R 7 Is optionally taken out ofSubstituted N-azetidinyl or optionally substituted N-morpholinyl. In some embodiments, two twins R 7 The groups are combined with the attached atoms to form an optionally substituted 4-to 10-membered heterocyclic group. In some embodiments, at least one R 7 is-OR 7A . In some embodiments, R 7A Is optionally substituted C 1-6 An alkyl group.
In some embodiments, n is 0.
In some embodiments, at least one R 7 Is cyclopropyl, 2-difluorocyclopropyl, difluoromethoxy, 2, 6-dimethylmorpholin-4-yl, N-azetidinyl, 3-fluorocyclobutyl, 2-methoxyethyl, ethoxy, methoxy, 2-difluoroethoxy, 2-difluoroethyl, trifluoromethyl, isopropyl, methyl, acetyl, fluoro, chloro, 1-methylpyrazol-3-yl, dimethylamino, N-methyl-N- (2-methoxyethyl) -amino, N-ethyl-N- (2-methoxyethyl) -amino, N- (2-propyl) -N- (2-methoxyethyl) -amino, 2-methoxyethylamino, 3-aza-8-oxa-bicyclo [4.3.0]Non-3-yl, 3-aza-7-oxa-bicyclo [4.3.0 ]Non-3-yl, 1-fluorocyclobutan-1-yl, 3-fluoropyrrolidin-1-yl, 3-methoxypyrrolidin-1-yl, oxetan-3-yl, N-methylindolin-4-yl, 2-difluoro-3-methylcyclopropan-1-yl, 3-methoxyazetidin-1-yl, 3-methoxypiperidin-1-yl, 1, 2-dimethyl-7-azaindol-4-yl, 1-methyl-7-azaindol-4-yl, 2, 3-methylenedioxyphenyl, N-methyl-N- (3-oxetan) amino, 3-oxetan-yloxy, 1-difluoro-5-azaspiro [2.3 ]]Hex-5-yl, 1-fluoromethyl-cyclopropyl, N- (3-tetrahydrofuranyl) methylamino, N-indolinyl, N-1, 4-oxaazepinyl, 2-fluoro-2-propyl, 1-difluoro-2-propyl, 2-difluoro-1-methylcyclopropan-1-yl, 1-methylcyclopropyl, 4-difluoropiperidin-1-yl, 2-methoxyethoxy, 3-difluorocyclobutan-1-yl N-methyl-N-1-methoxyprop-2-ylamino, 1-methoxyethyl, 4-methylpiperazinyl, 3-methylmorpholine, 2-difluoropropoxy, 3-methoxycyclobutyl, methylamino, 4-dimethylamino-3, 3-difluoropiperidinyl, 4-methylamino-3, 3-difluoropiperidinyl, 3-difluoropyrrolidinyl, N-methyl1-methylpyrazol-5-yl, 6-oxa-3-azabicyclo [3.1.1 ]Hept-3-yl, cyclopropoxy, 2, 6-dimethylpyridin-4-yl, 2-methylpyrrolidinyl, 4-oxabicyclo [4.1.0 ]]Hept-1-yl, N-methyl-N- (2, 6-dimethyltetrahydropyran-4-yl) amino or N-methyl-N-3-methyloxetan-3-ylmethylamino.
In some embodiments, R 1 Is hydrogen.
In another aspect, the invention provides compounds selected from compounds 1-308 of table 1A and pharmaceutically acceptable salts thereof.
TABLE 1A Compounds of the invention
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
In another aspect, the present invention provides compounds selected from compounds 309-856 in table 1B and pharmaceutically acceptable salts thereof.
TABLE 1B Compounds of the invention (cyclic and acyclic sulfones)
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
In some embodiments, the compound has a BRG1 IC of at least 5 50 And BRM IC 50 Is a ratio of (2). In some embodiments, the compounds have BRG1 IC 50 And BRM IC 50 At least 7. In some embodiments, the compounds have a BRG1 IC of at least 10 50 And BRM IC 50 Is a ratio of (2). In some embodiments, the compounds have a BRG1 IC of at least 15 50 And BRM IC 50 Is a ratio of (2). In some embodiments, the compounds have a BRG1 IC of at least 20 50 And BRM IC 50 Is a ratio of (2). In some embodiments, the compounds have a BRG1 IC of at least 25 50 And BRM IC 50 Is a ratio of (2). In some embodiments, the compounds have a BRG1IC of at least 30 50 And BRM IC 50 Is a ratio of (2).
In another aspect, the invention features a pharmaceutical composition that includes any of the compounds described above and a pharmaceutically acceptable excipient.
In another aspect, the invention features a method of reducing BAF complex activity in a cell, the method including contacting the cell with an effective amount of any of the above compounds or a pharmaceutical composition thereof.
In some embodiments, the cell is a cancer cell.
In another aspect, the invention features a method of treating a BAF complex-related disorder in a subject in need thereof, the method including administering to the subject an effective amount of any of the above compounds or a pharmaceutical composition thereof.
In some embodiments, the BAF complex-related disorder is cancer.
In another aspect, the invention features a method of inhibiting BRM, the method including contacting a cell with an effective amount of any of the above compounds or a pharmaceutical composition thereof. In some embodiments, the cell is a cancer cell.
In another aspect, the invention features a method of inhibiting BRG1, the method including contacting the cell with an effective amount of any of the above compounds or a pharmaceutical composition thereof. In some embodiments, the cell is a cancer cell.
In another aspect, the invention features a method of inhibiting BRM and BRG1, the method including contacting a cell with an effective amount of any of the above compounds or a pharmaceutical composition thereof. In some embodiments, the cell is a cancer cell.
In another aspect, the invention features a method of treating a disorder associated with a BRG1 loss-of-function mutation in a subject in need thereof, the method comprising administering to the subject an effective amount of any of the above compounds or pharmaceutical compositions thereof.
In some embodiments, the disorder associated with a BRG1 loss-of-function mutation is cancer. In other embodiments, the subject is determined to have a BRG1 loss of function disorder, e.g., is determined to have a BRG1 loss of function cancer (e.g., the cancer has been determined to include cancer cells having a BRG1 loss of function).
In another aspect, the invention features a method of inducing apoptosis in a cell, the method including contacting the cell with an effective amount of any of the above compounds or pharmaceutical compositions thereof. In some embodiments, the cell is a cancer cell.
In another aspect, a method of treating cancer in a subject in need thereof, the method comprising administering to the subject an effective amount of any of the above compounds or a pharmaceutical composition thereof.
In some embodiments of any of the above methods, the cancer is non-small cell lung cancer, colorectal cancer, bladder cancer, primary unknown cancer, glioma, breast cancer, melanoma, non-melanoma skin cancer, endometrial cancer, esophageal gastric cancer, pancreatic cancer, hepatobiliary tract cancer, soft tissue sarcoma, ovarian cancer, head and neck cancer, renal cell carcinoma, bone cancer, non-hodgkin lymphoma, small cell lung cancer, prostate cancer, embryonic tumor, blastoma, cervical cancer, thyroid cancer, salivary gland cancer, gastrointestinal neuroendocrine tumor, uterine sarcoma, gastrointestinal stromal tumor, CNS cancer, thymus tumor, adrenal cortex cancer, appendiceal cancer, small intestine cancer, or penile cancer.
In some embodiments of any of the above methods, the cancer is non-small cell lung cancer, colorectal cancer, bladder cancer, primary unknown cancer, glioma, breast cancer, melanoma, non-melanoma skin cancer, endometrial cancer, or penile cancer.
In some embodiments of any of the above methods, the cancer is a drug resistant cancer or is refractory to prior therapies (e.g., vemurafenib, dacarbazine, CTLA4 inhibitors, PD1 inhibitors, interferon therapy, BRAF inhibitors, MEK inhibitors, radiation therapy, temozolomide, irinotecan, CAR-T therapy, herceptin, paget-ta (perjeta), tamoxifen, hiloda (xeloda), docetaxel, platinum agents such as carboplatin, taxanes such as paclitaxel and docetaxel, ALK inhibitors, MET inhibitors, paclitaxel for injection (abaxane), Gemcitabine, algastine (avastin), eribulin mesylate (halaven), neratinib (neratinib), PARP inhibitors, ARN810, mTOR inhibitors, topotecan, gemzar, verat inhibitors, folate receptor antagonists, dimizumab, fosibirin (fosbretabulin), or PDL1 inhibitors.
In some embodiments of any of the foregoing methods, the cancer has or has been determined to have BRG1 mutation. In some embodiments of any of the foregoing methods, the BRG1 mutation is homozygous. In some embodiments of any of the foregoing methods, the cancer does not have, or has been determined to have, an Epidermal Growth Factor Receptor (EGFR) mutation. In some embodiments of any of the foregoing methods, the cancer does not have, or has been determined to have, anaplastic Lymphoma Kinase (ALK) driving mutations. In some embodiments of any of the foregoing methods, the cancer has or has been determined to have a KRAS mutation. In some embodiments of any of the foregoing methods, the BRG1 mutation is in the atpase catalytic domain of the protein. In some embodiments of any of the foregoing methods, BRG1 is mutated to be deleted at the C-terminus of BRG 1.
In another aspect, the present disclosure provides a method of treating a BAF-related disorder (e.g., cancer or viral infection) in a subject in need thereof. The method comprises contacting the cell with an effective amount of any of the above compounds or a pharmaceutically acceptable salt thereof or any of the above pharmaceutical compositions. In some embodiments, the disorder is a viral infection, such as a viral infection of the retrovirus family, e.g., lentivirus (e.g., human Immunodeficiency Virus (HIV) and delta retrovirus (e.g., human T-cell leukemia virus I (HTLV-I), human T-cell leukemia virus II (HTLV-II)), hepadnaviridae (Hepadnaviridae family) (e.g., hepatitis B Virus (HBV)), flaviviridae (e.g., hepatitis C Virus (HCV)), adenoviridae (e.g., human adenovirus), herpesviridae (e.g., human Cytomegalovirus (HCMV), E-B virus, herpes simplex virus 1 (HSV-1), herpes simplex virus 2 (HSV-2), human herpesvirus 6 (HHV-6), herpes virus K, CMV, varicella-zoster virus (variella-zoter virus)), papillomaviridae (e.g., human papilloma virus (HPV, e.g., HPV E1)), parvoviridae (e.g., parvoviridae B19), polyoviridae (e.g., JC virus and virus), paramoviridae (e.g., JC virus), schiffviridae (e.g., film, schnein) or in a multiple Rubella-37 virus (NF-37, a) or a multiple Rubella-37-virus condition implemented in the respiratory system (NF-37, NF-1, NF-35, or a multiple-duct-37-virus (scheme).
In another aspect, the present disclosure provides a method for treating a viral infection in a subject in need thereof. The method comprises administering to the subject an effective amount of any of the above compounds, or a pharmaceutically acceptable salt thereof, or any of the above pharmaceutical compositions. In some embodiments, the viral infection is a viral infection of the retrovirus family, such as lentivirus (e.g., human Immunodeficiency Virus (HIV) and delta retrovirus (e.g., human T-cell leukemia virus I (HTLV-I), human T-cell leukemia virus II (HTLV-II)), hepadnaviridae (e.g., hepatitis B Virus (HBV)), flaviviridae (e.g., hepatitis C Virus (HCV)), adenoviridae (e.g., human adenovirus), herpesviridae (e.g., human Cytomegalovirus (HCMV), E-B virus, herpes virus), herpes simplex virus 1 (HSV-1), herpes simplex virus 2 (HSV-2), human herpes virus 6 (HHV-6), herpes virus K, CMV, varicella-zoster virus), papillomaviridae (e.g., human papilloma virus (HPV, HPV E1)), parvoviridae (e.g., parvoviridae B19), polyomaviridae (e.g., JC virus and BK), paramyxoviridae (e.g., measles virus), or togaviridae (e.g., rubella virus).
In another aspect, the invention features a method of treating melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematological cancer in a subject in need thereof, the method comprising administering to the subject an effective amount of any of the above compounds or a pharmaceutical composition thereof.
In another aspect, the invention features a method of reducing tumor growth of melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematological cancer in a subject in need thereof, the method comprising administering to the subject an effective amount of any of the above compounds or a pharmaceutical composition thereof.
In another aspect, the invention features a method of inhibiting metastatic progression of melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematological cancer in a subject, the method comprising administering an effective amount of any of the above compounds or a pharmaceutical composition thereof.
In another aspect, the invention features a method of inhibiting metastatic colonization of melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematological cancer in a subject, the method comprising administering an effective amount of any of the foregoing compounds, or a pharmaceutical composition thereof.
In another aspect, the invention features a method of reducing the level and/or activity of BRG1 and/or BRM in a melanoma, prostate, breast, bone, renal cell or blood cancer cell, the method comprising contacting the cell with an effective amount of any of the above compounds or a pharmaceutical composition thereof.
In some embodiments of any one of the above aspects, the melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematological cancer cells are in a subject.
In some embodiments of any of the above aspects, an effective amount of a compound reduces the level and/or activity of BRG1 by at least 5% (e.g., 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%) relative to a reference. In some embodiments, an effective amount of a compound reduces the level and/or activity of BRG1 by at least 50% (e.g., 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%) relative to a reference. In some embodiments, an effective amount of a compound reduces the level and/or activity of BRG1 by at least 90% (e.g., 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%).
In some embodiments, an effective amount of a compound reduces the level and/or activity of BRG1 by at least 5% (e.g., 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%) relative to a reference for at least 12 hours (e.g., 14 hours, 16 hours, 18 hours, 20 hours, 22 hours, 24 hours, 30 hours, 36 hours, 48 hours, 72 hours, or more). In some embodiments, an effective amount of a compound reduces the level and/or activity of BRG1 by at least 5% (e.g., 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%) relative to a reference for at least 4 days (e.g., 5 days, 6 days, 7 days, 14 days, 28 days, or more).
In some embodiments of any of the above aspects, an effective amount of a compound reduces the level and/or activity of BRM by at least 5% (e.g., 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%) relative to a reference. In some embodiments, an effective amount of a compound reduces the level and/or activity of BRM by at least 50% (e.g., 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%) relative to a reference. In some embodiments, an effective amount of a compound reduces the level and/or activity of BRM by at least 90% (e.g., 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%).
In some embodiments, an effective amount of a compound reduces the level and/or activity of BRM by at least 5% (e.g., 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%) relative to a reference for at least 12 hours (e.g., 14 hours, 16 hours, 18 hours, 20 hours, 22 hours, 24 hours, 30 hours, 36 hours, 48 hours, 72 hours, or more). In some embodiments, an effective amount of a compound reduces the level and/or activity of BRM by at least 5% (e.g., 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%) relative to a reference for at least 4 days (e.g., 5 days, 6 days, 7 days, 14 days, 28 days, or more).
In some embodiments, the subject has cancer. In some embodiments, the cancer expresses BRG1 and/or BRM proteins and/or the cell or subject has been identified as expressing BRG1 and/or BRM. In some embodiments, the cancer expresses BRG1 protein and/or the cell or subject has been identified as expressing BRG1. In some embodiments, the cancer expresses BRM proteins and/or the cell or subject has been identified as expressing BRM. In some embodiments, the cancer is a melanoma (e.g., ocular pigment layer melanoma, mucosal melanoma, or cutaneous melanoma). In some embodiments, the cancer is prostate cancer. In some embodiments, the cancer is a hematologic cancer, such as multiple myeloma, large cell lymphoma, acute T-cell leukemia, acute myelogenous leukemia, myelodysplastic syndrome, immunoglobulin a lambda myeloma, diffuse mixed histiocyte and lymphocytic lymphoma, B-cell lymphoma, acute lymphoblastic leukemia (e.g., T-cell acute lymphoblastic leukemia or B-cell acute lymphoblastic leukemia), diffuse large cell lymphoma, or non-hodgkin's lymphoma. In some embodiments, the cancer is breast cancer (e.g., ER positive breast cancer, ER negative breast cancer, triple positive breast cancer, or triple negative breast cancer). In some embodiments, the cancer is bone cancer (e.g., ewing's sarcoma). In some embodiments, the cancer is a renal cell carcinoma (e.g., microphthalmia transcription factor (MITF) family translocated renal cell carcinoma (tRCC)). In some embodiments, the cancer is metastatic (e.g., the cancer has spread to the liver). Metastatic cancer may include cells that exhibit migration and/or invasion of migrating cells and/or include cells that exhibit endothelial recruitment and/or angiogenesis. In other embodiments, the metastatic cancer is a cell-migrating cancer. In other embodiments, the cell-migrating cancer is a non-metastatic cell-migrating cancer. The metastatic cancer may be a cancer that spreads by inoculating the surface of the peritoneal, pleural, pericardial or subarachnoid cavity. Alternatively, the metastatic cancer may be a cancer that spreads via the lymphatic system, or a cancer that spreads blood. In some embodiments, an effective amount of a compound of the invention is an amount effective to inhibit metastatic colonization of the liver by the cancer.
In some embodiments, the cancer carries mutations in GNAQ. In some embodiments, the cancer carries a mutation in GNA 11. In some embodiments, the cancer carries a mutation in PLCB 4. In some embodiments, the cancer carries a mutation in CYSLTR 2. In some embodiments, the cancer carries a mutation in BAP 1. In some embodiments, the cancer carries a mutation in SF3B 1. In some embodiments, the cancer carries a mutation in EIF1 AX. In some embodiments, the cancer carries TFE3 translocation. In some embodiments, the cancer carries TFEB translocation. In some embodiments, the cancer carries MITF translocation. In some embodiments, the cancer carries an EZH2 mutation. In some embodiments, the cancer carries a SUZ12 mutation. In some embodiments, the cancer carries an EED mutation.
In some embodiments of any of the above methods, the method further comprises administering an anti-cancer therapy to the subject or contacting the cells with an anti-cancer therapy, such as a chemotherapeutic or cytotoxic agent, immunotherapy, surgery, radiation therapy, thermal therapy, or photocoagulation therapy, or a combination thereof. In some embodiments, the anti-cancer therapy is a chemotherapeutic or cytotoxic agent, such as an antimetabolite, an antimitotic, an antitumor antibiotic, an asparagine-specific enzyme, a bisphosphonate, an antitumor agent, an alkylating agent, a DNA-repair enzyme inhibitor, a histone deacetylase inhibitor, a corticosteroid, a demethylating agent, an immunomodulator, a janus-related kinase inhibitor, a phosphatidylinositol 3-kinase (phospho-kinase) inhibitor, a proteasome inhibitor, or a tyrosine kinase inhibitor, or a combination thereof.
In some embodiments of any of the above methods, the compounds of the invention are used in combination with another anti-cancer therapy, such as surgery, a MEK inhibitor, and/or a PKC inhibitor, for treating ocular pigment layer melanoma. For example, in some embodiments, the method further comprises performing surgery before, after, or simultaneously with the administration of the compounds of the invention. In some embodiments, the method further comprises administering a MEK inhibitor and/or a PKC inhibitor prior to, after, or concurrently with administration of the compound of the invention.
In some embodiments, the anti-cancer therapy and the compound of the invention are administered within 28 days of each other and each in an amount effective together to treat the subject.
In some embodiments, the subject or cancer has and/or has been identified as having a BRG1 loss-of-function mutation.
In some embodiments, the cancer is resistant to one or more chemotherapeutic or cytotoxic agents (e.g., by gene markers, the cancer has been determined to be resistant to a chemotherapeutic or cytotoxic agent, or may be resistant to a chemotherapeutic or cytotoxic agent, e.g., a cancer that does not respond to a chemotherapeutic or cytotoxic agent). In some embodiments, the cancer is not responsive to one or more chemotherapeutic agents or cytotoxic agents. In some embodiments, the cancer is resistant or non-responsive to dacarbazine, temozolomide, cisplatin, trosoxidan (treosulfan), fotemustine, IMCgp100, CTLA-4 inhibitors (e.g., time ipilimumab), PD-1 inhibitors (e.g., nivolumab or pembrolizumab (pembrolizumab)), PD-L1 inhibitors (e.g., atizumab (atezolizumab), avistuzumab (avelumab) or dulciton You Shan (durvalumab)), mitogen-activated protein kinase (MEK) inhibitors (e.g., semmetinib), bimetainib (binimetinib) or trimetinib (tametib)), and/or Protein Kinase C (PKC) inhibitors (e.g., sotatastin Qu Tuolin (sotalorin) or IDE 196).
In some embodiments, the cancer is resistant to or unable to respond to previously administered therapeutic agents, such as MEK inhibitors or PKC inhibitors, for treating ocular pigment layer melanoma. For example, in some embodiments, the cancer is resistant or non-responsive to a mitogen-activated protein kinase (MEK) inhibitor (e.g., semanteme, bimatinib, or trimetinib) and/or a Protein Kinase C (PKC) inhibitor (e.g., cord Qu Tuolin or IDE 196).
Chemical terminology
The terminology used herein is for the purpose of describing particular embodiments and is not intended to be limiting.
For any of the following chemical definitions, the numbers following the atomic symbols represent the total number of atoms of the element present in the particular chemical moiety. As will be appreciated, other atoms as described herein, such as H atoms or substituents, may be present as desired to satisfy the valences of the atoms. For example, unsubstituted C 2 Alkyl has the formula-CH 2 CH 3 . When used with the groups defined herein, reference to the number of carbon atoms includes the carbs in the acetal and ketal groups, but does not include the carbonyl carbons in the acyl, ester, carbonate or carbamate groups. References to the number of oxygen, nitrogen or sulfur atoms in a heteroaryl group include only those atoms that form part of a heterocyclic ring.
As used herein, the term "acyl" refers to H or alkyl attached to a parent molecular group through a carbonyl group as defined herein, and is exemplified by formyl (i.e., carboxyaldehyde), acetyl, trifluoroacetyl, propionyl, and butyryl. Exemplary unsubstituted acyl groups include 1 to 6, 1 to 11, or 1 to 21 carbons.
As used herein, the term "alkenyl" refers to a branched or straight chain monovalent saturated aliphatic hydrocarbon group having 2 to 20 carbon atoms (e.g., 2 to 16 carbon atoms, 2 to 10 carbon atoms, or 2 to 6 carbon atoms). Alkenyl groups may be monovalent or multivalent, for example. Those skilled in the art will recognize from this context the number of available valences.
As used herein, the term "alkyl" refers to branched or straight chain monovalent saturated aliphatic hydrocarbon groups having 1 to 20 carbon atoms (e.g., 1 to 16 carbon atoms, 1 to 10 carbon atoms, and 1 to 6 carbon atoms, or 1 to 3 carbon atoms). The alkyl group may be, for example, monovalent or multivalent. Those skilled in the art will recognize from this context the number of available prices.
The term "amino" as used herein means-N (R N1 ) 2 Wherein each R is N1 Is independently H, OH, NO 2 、N(R N2 ) 2 、SO 2 OR N2 、SO 2 R N2 、SOR N2 An N-protecting group, alkyl, alkoxy, aryl, aralkyl, cycloalkyl, acyl (e.g., acetyl, trifluoroacetyl or other groups described herein), heteroaryl, or heterocyclyl, wherein each of these list R N1 The groups may be optionally substituted; or two R N1 Combined with the atoms to which they are attached to form a heterocyclic or heteroaryl group, and wherein each R N2 Independently is H, alkyl or aryl. The amino group of the present invention may be an unsubstituted amino group (i.e., -NH- 2 ) Or substituted amino (i.e. -N (R) N1 ) 2 )。
As used herein, the term "aryl" refers to an aromatic mono-or multi-carbocyclic group of 6 to 12 carbon atoms having at least one aromatic ring. Examples of such groups include, but are not limited to, phenyl, naphthyl, 1,2,3, 4-tetrahydronaphthyl, 1, 2-dihydronaphthyl, indanyl, and 1H-indenyl. Aryl groups may be monovalent or multivalent, for example. Those skilled in the art will recognize from this context the number of available prices.
The term "arylalkyl" as used herein means substituted with aryl groupsIs a hydrocarbon group. Exemplary unsubstituted arylalkyl groups are 7 to 30 carbons (e.g., 7 to 16 or 7 to 20 carbons, such as C 1 -C 6 Alkyl C 6 -C 10 Aryl, C 1 -C 10 Alkyl C 6 -C 10 Aryl or C 1 -C 20 Alkyl C 6 -C 10 Aryl), such as benzyl and phenethyl. In some embodiments, each of the alkyl and aryl groups may be further substituted with 1,2,3, or 4 substituents as defined herein for the corresponding group.
The term "azido" as used herein means-N 3 A group.
As used herein, the term "bridged polycycloalkyl" refers to a bridged polycycloalkyl group of 5 to 20 carbons that contains 1 to 3 bridges.
As used herein, the term "cyano" represents a —cn group.
As used herein, the term "carbocyclyl" refers to a non-aromatic C 3 -C 12 A monocyclic, bicyclic or tricyclic structure wherein the rings are formed by carbon atoms. Carbocyclyl structures include cycloalkyl and unsaturated carbocyclyl.
As used herein, the term "cycloalkyl" refers to saturated, non-aromatic and monovalent mono-or multicyclic groups of 3 to 10, preferably 3 to 6 carbon atoms. The term is further exemplified by groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl and adamantyl. Cycloalkyl groups may be, for example, monovalent or multivalent. Those skilled in the art will recognize from this context the number of available valences.
As used herein, the term "halogen" refers to a fluoro (fluoro), chloro (chloro), bromo (bromo) or iodo (iodo) group.
As used herein, the term "heteroalkyl" refers to an alkyl group as defined herein wherein one or more of the constituent carbon atoms have been replaced with nitrogen, oxygen, or sulfur. In some embodiments, the heteroalkyl group may be further substituted with 1, 2, 3, or 4 substituents as described herein for the alkyl group. Examples of heteroalkyl are "alkoxy," as used herein, refers to alkyl-O- (e.g., methoxy and ethoxy). Heteroalkyl groups may be, for example, monovalent or multivalent. Those skilled in the art will recognize from the context the number of available valences.
As used herein, the term "heteroaryl" refers to a mono-or polycyclic group of 5-14 (e.g., 5 to 12 or 5 to 10) atoms having at least one aromatic ring and containing 1, 2, or 3 ring atoms selected from nitrogen, oxygen, and sulfur, the remaining ring atoms being carbon. In some embodiments, heteroaryl is C 1 -C 9 Heteroaryl (e.g. C 2 -C 9 Heteroaryl). One or both ring carbon atoms of the heteroaryl group may be substituted with a carbonyl group. Examples of heteroaryl groups are pyridyl, pyrazolyl, benzoxazolyl, benzimidazolyl, benzothiazolyl, imidazolyl, oxazolyl, thiazolyl, benzomorpholinyl, benzopiperidinyl and indolinyl. Heteroaryl groups may be monovalent or multivalent, for example. Those skilled in the art will recognize from this context the number of available valences.
As used herein, the term "heteroarylalkyl" means an alkyl group substituted with a heteroaryl group. Exemplary unsubstituted heteroaralkyl groups are 7 to 30 carbons (e.g., 7 to 16 or 7 to 20 carbons, such as C 2 -C 9 Heteroaryl C 1 -C 6 Alkyl, C 2 -C 9 Heteroaryl C 1 -C 10 Alkyl or C 2 -C 9 Heteroaryl C 1 -C 20 Alkyl). In some embodiments, each of the alkyl and heteroaryl groups may be further substituted with 1, 2, 3, or 4 substituents as defined herein for the corresponding group.
As used herein, the term "heterocyclyl" refers to a mono-or polycyclic group having from 3 to 14 (e.g., 4 to 12) atoms having at least one ring containing 1, 2, 3, or 4 ring atoms selected from N, O or S, wherein none of the rings is aromatic. In some embodiments, the heterocyclyl is C 2 -C 9 A heterocyclic group. Examples of heterocyclyl groups include, but are not limited to, morpholinyl, thiomorpholinyl, piperazinyl, piperidinyl, pyranyl, pyrrolidinyl, tetrahydropyranyl, tetrahydrofuranyl, 1, 3-dioxanyl, aza-oxybicyclo [4.3.0]Nonyl and aza-oxy bicyclo [4.4.0]And (3) a decyl group. The heterocyclic group may be, for example, monovalent or multivalent. Those skilled in the artThe person will recognize from the context the number of available valences.
As used herein, the term "heterocyclylalkyl" refers to an alkyl group substituted with a heterocyclyl group. Exemplary unsubstituted heterocycloalkyl groups are 7 to 30 carbons (e.g., 7 to 16 or 7 to 20 carbons, such as C 2 -C 9 Heterocyclyl C 1 -C 6 Alkyl, C 2 -C 9 Heterocyclyl C 1 -C 10 Alkyl or C 2 -C 9 Heterocyclyl C 1 -C 20 Alkyl). In some embodiments, each of the alkyl and heterocyclyl groups may be further substituted with 1, 2, 3, or 4 substituents as defined herein for the corresponding group.
As used herein, the term "hydroxyalkyl" refers to an alkyl group in which the alkyl group is substituted with an-OH group.
As used herein, the term "hydroxy" means an-OH group.
As used herein, the term "N-protecting group" means those groups intended to protect an amino group from undesired reactions during the synthetic process. A commonly used N-protecting group is disclosed in Greene, "Protective Groups in Organic Synthesis," 3 rd edition (John Wiley & Sons, new York, 1999). N-protecting groups include, but are not limited to, acyl, aroyl or carbamoyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthaloyl, o-nitrophenoxyacetyl, α -chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl and chiral auxiliary such as protected or unprotected D, L or D, L-amino acids such as alanine, leucine and phenylalanine; sulfonyl-containing groups such as benzenesulfonyl and p-toluenesulfonyl; urethane forming groups such as benzyloxycarbonyl, p-chlorobenzoxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2-nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3, 4-dimethoxybenzyloxycarbonyl, 3, 5-dimethoxybenzyloxycarbonyl, 2,4-20 dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2-nitro-4, 5-dimethoxybenzyloxycarbonyl, 3,4, 5-trimethoxybenzyloxycarbonyl, 1- (p-biphenyl) -1-methylethoxycarbonyl,. Alpha., α -dimethyl-3, 5-dimethoxybenzyloxycarbonyl, benzhydryl, t-butoxycarbonyl, diisopropylmethoxycarbonyl, isopropoxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2, -trichloroethoxycarbonyl, phenoxycarbonyl, 4-nitrophenoxycarbonyl, fluorenyl-9-methoxycarbonyl, cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl and phenylthiocarbonyl (phenylthiocarbonyl), arylalkyl groups such as benzyl, triphenylmethyl and benzyloxymethyl, and silyl groups such as trimethylsilyl. Preferred N-protecting groups are alloc, formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, benzenesulfonyl, benzyl, t-butoxycarbonyl (Boc) and benzyloxycarbonyl (Cbz).
The term "nitro" as used herein means-NO 2 A group.
The term "sulfhydryl" as used herein means a-SH group.
The alkyl, heteroalkyl, carbocyclyl (e.g., cycloalkyl), aryl, heteroaryl, and heterocyclyl groups may be substituted or unsubstituted. When substituted, 1 to 4 substituents are typically present unless otherwise indicated. Substituents include, for example: alkyl (e.g., unsubstituted and substituted, wherein the substituents include any of the groups described herein, e.g., aryl, halogen, hydroxy), aryl (e.g., substituted and unsubstituted phenyl), carbocyclyl (e.g., substituted and unsubstituted cycloalkyl), halogen (e.g., fluoro), hydroxy, heteroalkyl (e.g., substituted and unsubstituted methoxy, ethoxy, or thioalkoxy), heteroaryl, heterocyclyl, amino (e.g., NH) 2 Or mono-or dialkylamino), azido, cyano, nitro or sulfhydryl. Aryl, carbocyclyl (e.g., cycloalkyl), heteroaryl, and heterocyclyl groups may also be substituted with alkyl groups (unsubstituted and substituted, e.g., arylalkyl groups (e.g., substituted and unsubstituted benzyl)).
The compounds of the invention may have one or more asymmetric carbon atoms and may exist as optically pure enantiomers, mixtures of enantiomers such as racemates, optically pure diastereomers, mixtures of diastereomers, diastereoisomeric racemates or mixtures of diastereoisomeric racemates. Optically active forms can be obtained, for example, by resolution of the racemate, by asymmetric synthesis or asymmetric chromatography (chromatography using chiral adsorbents or eluents). That is, certain disclosed compounds may exist in various stereoisomeric forms. Stereoisomers are compounds that differ only in their spatial arrangement. Enantiomers are mirror image non-overlapping pairs of stereoisomers, most commonly because they contain asymmetrically substituted carbon atoms as chiral centers. "enantiomer" means one of a pair of molecules that are mirror images of each other and that are non-overlapping. Diastereomers are stereoisomers that are not mirror-image related, most commonly because they contain two or more asymmetrically substituted carbon atoms and represent the configuration of substituents around one or more chiral carbon atoms. Enantiomers of a compound may be prepared, for example, by separating the enantiomer from the racemate using one or more well-known techniques and methods, such as chiral chromatography and separation methods based thereon. One skilled in the art can readily determine the appropriate techniques and/or methods for separating enantiomers of compounds described herein from a racemic mixture. "racemate" or "racemic mixture" refers to a compound comprising two enantiomers, wherein such mixture does not exhibit optical activity; i.e. they do not rotate the plane of polarized light. "geometric isomer" refers to an isomer in which the orientation of the substituent atoms relative to the carbon-carbon double bond, cycloalkyl ring, or bridged bicyclic ring system is different. The atoms on each side of the carbon-carbon double bond (except H) may be in the E (substituents on opposite sides of the carbon-carbon double bond) or Z (substituents oriented on the same side) configuration. "R", "S", "R", "E", "Z", "cis" and "trans" refer to configuration relative to the core molecule. Certain disclosed compounds may exist in atropisomeric forms. Atropisomers are stereoisomers produced by a hindered rotation about a single bond, where the spatial strain barrier of the rotation is high enough to allow separation of conformational isomers. The compounds of the invention may be prepared as individual isomers by isomer-specific synthesis or resolution from mixtures of isomers. Conventional resolution techniques include formation of a salt of the free base of each isomer of the isomer pair using an optically active acid (followed by fractional crystallization and regeneration of the free base), formation of a salt of the acid form of each isomer of the isomer pair using an optically active amine (followed by fractional crystallization and regeneration of the free acid), formation of an ester or amide of each isomer of the isomer pair using an optically pure acid, amine or alcohol (followed by chromatographic separation and removal of chiral auxiliary), or resolution of an isomer mixture of the starting material or end product using various well known chromatographic methods. When the stereochemistry of a disclosed compound is named or described by structure, the named or described stereoisomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% by weight relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% by weight optically pure. When a single diastereomer is named or depicted by structure, the depicted or named diastereomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% pure by weight. The percent optical purity is the ratio of the weight of an enantiomer to the weight of the enantiomer plus the weight of its optical isomer. The diastereomeric purity by weight is the ratio of the weight of one diastereomer to the weight of all diastereomers. When the stereochemistry of a disclosed compound is named or described by structure, the named or described stereoisomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% mole fraction pure relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% mole fraction pure. When a single diastereomer is named or described by structure, the diastereomer described or named is at least 60%, 70%, 80%, 90%, 99% or 99.9% mole fraction pure. The percent purity in mole fraction is the ratio of the moles of enantiomer to the moles of enantiomer plus the moles of its optical isomer. Similarly, the mole fraction purity percentage is the ratio of the moles of diastereomers to the moles of diastereomers plus the moles of isomers thereof. When a disclosed compound is named or described by structure without indicating stereochemistry and the compound has at least one chiral center, it is to be understood that the name or structure encompasses the enantiomer of the compound that does not contain the corresponding optical isomer, the racemic mixture of the compound, or a mixture enriched in one enantiomer relative to its corresponding optical isomer. When a disclosed compound is named or described by structure without indicating stereochemistry and has two or more chiral centers, it is to be understood that the name or structure encompasses diastereomers which are free of other diastereomers, numerous diastereomers which are free of other diastereomers, mixtures of diastereomers in which one diastereomer is enriched relative to the other diastereomers, or mixtures of diastereomers in which one or more diastereomers are enriched relative to the other diastereomers. The present invention includes all such forms.
The compounds of the present disclosure also include all isotopes of atoms present in the intermediates or final compounds. "isotope" refers to atoms of different mass numbers having the same atomic number but produced by different numbers of neutrons in the core. Isotopes of hydrogen include, for example, tritium and deuterium.
Unless otherwise indicated, structures depicted herein are also intended to include compounds that differ only in the presence of one or more isotopically enriched atoms. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, for example 2 H、 3 H、 11 C、 13 C、 14 C、 13 N、 15 N、 15 O、 17 O、 18 O、 32 P、 33 P、 35 S、 18 F、 36 Cl、 123 I and 125 I. isotopically-labeled compounds (e.g 3 H and 14 c-labeled) can be used in compound or substrate tissue distribution assays. Tritiated (i.e. tritiated) 3 H) And carbon-14 (i.e 14 C) Isotopes may be useful because of their ease of preparation and detectability. In addition, substitution of heavier isotopes such as deuterium (i.e 2 H) Certain therapeutic advantages (e.g., increased in vivo half-life or reduced dosage requirements) may be provided due to greater metabolic stability. In some embodiments, one or more hydrogen atoms are replaced with 2 H or 3 H being substituted for or one or more carbon atoms being substituted 13 C-or 14 C-enriched carbon substitution. Positron emitting isotopes, e.g 15 O、 13 N、 11 C and C 18 F can be used in Positron Emission Tomography (PET) studies to examine substrate receptor occupancy. The preparation of isotopically-labeled compounds is known to those skilled in the art. For example, isotopically-labeled compounds can generally be prepared by following procedures analogous to those disclosed for compounds of the application described herein by substituting an isotopically-labeled reagent for a non-isotopically-labeled reagent.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this application belongs. Methods and materials for the present disclosure are described herein; other suitable methods and materials known in the art may also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.
Definition of the definition
In the present application, unless the context clearly indicates otherwise, (i) the term "a" or (an) "may be understood to mean" at least one "; (ii) the term "or" is understood to mean "and/or"; and (iii) the terms "comprising" and "including" may be understood to encompass the listed components or steps on a per-item basis, whether presented alone or with one or more additional components or steps.
As used herein, the terms "about" and "approximately" refer to values within 10% above or below the stated value. For example, the term "about 5nM" means a range of 4.5 to 5.5 nM.
As used herein, the term "administering" refers to administering a composition (e.g., a compound or formulation comprising a compound as described herein) to a subject or system. Administration to an animal subject (e.g., to a human) may be by any suitable route. For example, in some embodiments, administration may be bronchial (including by bronchial instillation), buccal, enteral, intradermal, intraarterial, intradermal, intragastric, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intratumoral, intravenous, intraventricular, mucosal, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (including by intratracheal instillation), transdermal, vaginal, and vitreous.
As used herein, the term "BAF complex" refers to BRG1 or HRBM-related factor complex in human cells.
As used herein, the term "BAF complex phase Guan Bingzheng" refers to a condition caused or affected by the activity level of the BAF complex.
As used herein, the term "BRG1 loss-of-function mutation" refers to a mutation in BRG1 that results in a decrease in protein activity (e.g., a decrease in BRG1 activity of at least 1%, e.g., a decrease in BRG1 activity of 2%, 5%, 10%, 25%, 50%, or 100%). Exemplary BRG1 loss-of-function mutations include, but are not limited to, homozygous BRG1 mutations and deletions at the C-terminus of BRG 1.
As used herein, the term "BRG1 loss of function disorder" refers to a disorder that exhibits reduced BRG1 activity (e.g., reduced BRG1 activity by at least 1%, e.g., reduced BRG1 activity by 2%, 5%, 10%, 25%, 50% or 100%).
The term "cancer" refers to a condition caused by proliferation of malignant tumor cells, such as tumors, neoplasms, carcinomas, sarcomas, leukemias, and lymphomas.
As used herein, "combination therapy" or "administration in combination" refers to administration of two (or more) different agents or treatments to a subject as part of a defined treatment regimen for a particular disease or disorder. The treatment regimen defines the dose and administration period of each agent such that the effects of the individual agents on the subject overlap. In some embodiments, delivery of two or more agents is simultaneous or concurrent, and the agents may be co-formulated. In some embodiments, the two or more agents are not co-formulated and are administered in a sequential manner as part of a prescription regimen. In some embodiments, the combined administration of two or more agents or treatments results in a reduction in symptoms or other parameters associated with the disorder that is greater than would be observed if the other agent or treatment was delivered alone or in the absence of the other agent. The effects of both treatments may be partially additive, fully additive, or greater than additive (e.g., synergistic). Sequential or substantially simultaneous administration of each therapeutic agent may be accomplished by any suitable route including, but not limited to, oral route, intravenous route, intramuscular route, and direct absorption through mucosal tissue. The therapeutic agents may be administered by the same route or by different routes. For example, a first therapeutic agent in combination may be administered by intravenous injection, while a second therapeutic agent in combination may be administered orally.
By "determining the level of a protein or RNA" is meant the direct or indirect detection of a protein or RNA by methods known in the art. "directly determining" refers to performing a process (e.g., an assay or test on a sample or the term "analyzing a sample" as defined herein) to obtain a physical entity or value. "indirect determination" refers to receiving a physical entity or value from another party or source (e.g., a third party laboratory that directly obtains the physical entity or value). Methods of measuring protein levels generally include, but are not limited to, western blotting, immunoblotting, enzyme-linked immunosorbent assays (ELISA), radioimmunoassays (RIA), immunoprecipitation, immunofluorescence, surface plasmon resonance, chemiluminescence, fluorescence polarization, phosphorescence, immunohistochemical analysis, matrix assisted laser desorption/ionization time of flight (MALDI-TOF) mass spectrometry, liquid Chromatography (LC) -mass spectrometry, microcytosis, microscopy, fluorescence Activated Cell Sorting (FACS), and flow cytometry, as well as assays based on protein properties, including but not limited to enzymatic activity or interactions with other protein partners. Methods for measuring RNA levels are known in the art and include, but are not limited to, quantitative polymerase chain reaction (qPCR) and Northern blot analysis.
By "reduced level" or "increased level" of protein or RNA is meant that the protein or RNA level is reduced or increased, respectively, by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500% or more, by greater than about 10%, about 15%, about 20%, about 50%, about 75%, about 100% or about 200%, by less than about 0.01-fold, about 0.02-fold, about 0.1-fold, about 0.3-fold, about 0.5-fold, about 0.8-fold or less, or by greater than about 1.2-fold, about 1.4-fold, about 1.5-fold, about 1.8-fold, about 2-fold, about 3.0-fold, about 5-fold, about 0.3-fold, about 5-fold, about 0.5-fold, about 0.1.3-fold, about 5-fold, or about 100% or more than the reference. Protein levels are expressed in mass/vol (e.g., g/dL, mg/mL, μg/mL, ng/mL) or as mass or percentage relative to the total protein in the sample.
By "reducing the activity of BAF complexes" is meant reducing the level of activity associated with BAF complexes or related downstream effects. A non-limiting example of reducing BAF complex activity is Sox2 activation. The activity level of the BAF complex may be measured using any method known in the art, for example, the method described in Kadoch et al Cell,2013,153,71-85, which method is incorporated herein by reference.
As used herein, the term "inhibiting BRM" refers to blocking or reducing the level or activity of an atpase catalytic binding domain or bromodomain of a protein. BRM inhibition may be determined using methods known in the art, such as BRM atpase assay, nano DSF assay, or BRM luciferase cell assay.
As used herein, the term "LXS196", also referred to as IDE196, refers to PKC inhibitors having the structure:
or a pharmaceutically acceptable salt thereof.
As used herein, the term "pharmaceutical composition" means a composition comprising a compound described herein formulated with pharmaceutically acceptable excipients and suitable for administration to a mammal, such as a human. Typically, the pharmaceutical compositions are manufactured or sold under the approval of government regulatory authorities as part of a therapeutic regimen for the treatment of a disease in a mammal. The pharmaceutical composition may be formulated, for example, for oral administration in unit dosage form (e.g., tablet, capsule, caplet, gel cap, or syrup); for topical administration (e.g., as a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use); or in any other pharmaceutically acceptable formulation.
As used herein, "pharmaceutically acceptable excipient" refers to any ingredient other than the compounds described herein (e.g., a vehicle capable of suspending or dissolving the active compound) and having properties that are substantially non-toxic and non-inflammatory in the patient. Excipients may include, for example: anti-adherent agents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colorants), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavoring agents, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, adsorbents, suspending or dispersing agents, sweeteners, and hydration water.
As used herein, the term "pharmaceutically acceptable salt" refers to any pharmaceutically acceptable salt of a compound, e.g., any compound of formula I. Pharmaceutically acceptable salts of any of the compounds described herein can include those suitable for use in contact with human and animal tissue without undue toxicity, irritation, allergic response, and commensurate with a reasonable benefit/risk ratio, within the scope of sound medical judgment. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: berge et al, J.pharmaceutical Sciences 66:1-19,1977 and Pharmaceutical Salts: properties, selection, and Use, (eds. P.H.Stahl and C.G.Wermuth), wiley-VCH,2008. Salts may be prepared in situ during the final isolation and purification of the compounds described herein, or separately by reacting the free base groups with a suitable organic acid.
The compounds of the present invention may have an ionizable group so as to be capable of being prepared as a pharmaceutically acceptable salt. These salts may be acid addition salts involving inorganic or organic acids, or in the case of the acidic form of the compounds of the invention, the salts may be prepared from inorganic or organic bases. Typically, the compounds are prepared or used as pharmaceutically acceptable salts, which are prepared as addition products of pharmaceutically acceptable acids or bases. Suitable pharmaceutically acceptable acids and bases and methods for preparing suitable salts are well known in the art. Salts may be prepared from pharmaceutically acceptable non-toxic acids and bases, including inorganic and organic acids and bases.
By "reference" is meant any useful reference for comparing protein or RNA levels. The reference may be any sample, standard curve or level used for comparison purposes. The reference may be a normal reference sample or a reference standard or level. The "reference sample" may be, for example, a control, e.g., a predetermined negative control value, e.g., a "normal control" or a previous sample taken from the same subject; a sample from a normal healthy subject, such as normal cells or normal tissue; a sample (e.g., a cell or tissue) from a subject not suffering from a disease; a sample from a subject diagnosed with a disease but not yet treated with a compound of the invention; a sample from a subject who has been treated with a compound of the invention; or a sample of purified protein or RNA (e.g., any of those described herein) at a known normal concentration. By "reference standard or level" is meant a value or number derived from a reference sample. A "normal control value" is a predetermined value that is indicative of a non-disease state, such as that expected in healthy control subjects. Typically, the normal control value is expressed as a range ("between X and Y"), a high threshold ("no higher than X") or a low threshold ("no lower than X"). Subjects with a measured value that is within the normal control value for a particular biomarker are typically referred to as "within normal limits" for that biomarker. A normal reference standard or level may be a value or number derived from a normal subject not suffering from a disease or disorder (e.g., cancer); a subject that has been treated with a compound of the invention. In preferred embodiments, the reference sample, standard or level is matched to the sample of the sample subject by at least one of the following criteria: age, weight, sex, stage of disease and general health. Standard curves of levels of purified protein or RNA (e.g., any of the proteins or RNAs described herein) within normal reference ranges may also be used as references.
As used herein, the term "subject" refers to any organism to which a composition according to the invention may be administered, e.g., for experimental, diagnostic, prophylactic and/or therapeutic purposes. Typical subjects include any animal (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans). The subject may seek or need treatment, require treatment, be receiving treatment, or be a person or animal under the care of a trained professional for a particular disease or condition.
As used herein, the term "treatment" or "treatment" refers to therapeutic treatment or any measure that aims to slow down (alleviate) an undesired physiological condition, disorder or disease, or to obtain a beneficial or desired clinical result. Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; a reduction in the extent of a condition, disorder or disease; a stable (i.e., non-worsening) state of a condition, disorder or disease; delay of onset or slowing of progression of the condition, disorder or disease; improvement or alleviation of a condition, disorder, or disease state (whether partial or complete); an improvement in at least one measurable physical parameter that is not necessarily discernible by the patient; or enhancement or amelioration of a condition, disorder or disease. Treatment involves eliciting a clinically significant response without undue levels of side effects. Treatment also includes prolonged survival compared to the expected survival if not treated. The compounds of the invention may also be used for "prophylactic treatment" or "prophylaxis" of a disorder, e.g., in a subject at increased risk of developing a disorder.
As used herein, the terms "variant" and "derivative" are used interchangeably and refer to naturally occurring, synthetic, and semisynthetic analogs of a compound, peptide, protein, or other substance described herein. Variants or derivatives of the compounds, peptides, proteins, or other substances described herein may retain or improve the biological activity of the starting material.
The details of one or more embodiments of the invention are set forth in the description below. Other features, objects, and advantages of the invention will be apparent from the description and claims.
Brief Description of Drawings
FIG. 1 is a schematic diagram illustrating inhibition of cell proliferation of several cancer cell lines by BRG1/BRM inhibitors (Compound A).
FIG. 2A is a schematic diagram illustrating inhibition of cell proliferation of an eye-layer melanoma cell line 92-1 by BRG1/BRM inhibitor (Compound A), MEK inhibitor (Semtinib), and PKC inhibitor (LXS 196).
FIG. 2B is a schematic diagram illustrating inhibition of cell proliferation of the uveal melanoma cell line MP41 by BRG1/BRM inhibitors (Compound A), MEK inhibitors (Semtinib), and PKC inhibitors (LXS 196).
FIG. 3 is a schematic diagram illustrating inhibition of cell proliferation of several cancer cell lines by BRG1/BRM inhibitors (Compound B).
FIG. 4 is a graph illustrating area under the curve (AUC) calculated from the dose-response curve of a cancer cell line treated with a BRG1/BRM inhibitor.
FIG. 5 is a schematic diagram illustrating inhibition of cell proliferation of uveal melanoma and non-small cell lung cancer cell lines by BRG1/BRM inhibitors (Compound B).
FIG. 6A is a schematic diagram illustrating inhibition of cell proliferation of an eye-layer melanoma cell line 92-1 by BRG1/BRM inhibitor (Compound B), MEK inhibitor (Semtinib), and PKC inhibitor (LXS 196).
FIG. 6B is a schematic diagram illustrating inhibition of cell proliferation of the uveal melanoma cell line MP41 by BRG1/BRM inhibitors (Compound B), MEK inhibitors (Semtinib), and PKC inhibitors (LXS 196).
Fig. 7A is a schematic diagram illustrating inhibition of cell proliferation by a PKC inhibitor (LXS 196) of a parental and PKC inhibitor refractory eye pigment layer melanoma cell line.
FIG. 7B is a schematic diagram illustrating inhibition of cell proliferation by BRG1/BRM inhibitors (Compound B) of parent and PKC inhibitor refractory uveal melanoma cell lines.
FIG. 8A is a schematic representation of the inhibition of tumor growth by an exemplary BRG1/BRM inhibitor (Compound C) in mice transplanted with an eye pigment layer melanoma cell line.
FIG. 8B is a schematic representation of tumor size of mice exemplified with an engrafted uveal melanoma cell line and administered a BRG1/BRM inhibitor (Compound C).
FIG. 8C is a schematic diagram illustrating weight changes in mice transplanted with an eye pigmentation layer melanoma cell line and administered a BRG1/BRM inhibitor (Compound C).
Detailed Description
The present disclosure features compounds for inhibiting BRG1 and optionally BRG 1. These compounds may be used to modulate BAF complex activity, for example, for the treatment of BAF-related disorders, such as cancer (e.g., BRG 1-loss of function disorders). Exemplary compounds described herein include compounds having the structure of formula I:
wherein the method comprises the steps of
m is 0, 1, 2 or 3;
n is 0, 1, 2, 3 or 4;
X 1 is-S-, -SO 2 -or-S (O) (NH) -;
X 2 is N or CR 8
R 1 Is hydrogen or optionally substitutedC of (2) 1 -C 6 An alkyl group;
each R 2 And each R 3 Independently hydrogen, optionally substituted C 1 -C 6 Alkyl or optionally substituted C 1 -C 6 A heteroalkyl group;
L 1 is an optionally substituted 9-to 10-membered bicyclic heterocyclyl or an optionally substituted 9-to 10-membered bicyclic heteroaryl;
L 2 absent, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted C 6 -C 10 Aryl, optionally substituted 5-to 14-membered heteroaryl, or optionally substituted 4-to 14-membered heterocyclyl;
R 4 is hydrogen, halogen, optionally substituted C 1 -C 6 Alkyl or optionally substituted C 3 -C 10 Cycloalkyl;
R 5 is optionally substituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl or optionally substituted amino, and R 6 Is hydrogen, halogen, cyano, optionally substituted C 1 -C 6 Alkyl, optionally substituted C 2 -C 6 Alkenyl or optionally substituted C 3 -C 10 Cycloalkyl; or R is 5 And R is 6 And together with the attached atoms, are combined into an optionally substituted 5-to 8-membered heterocyclyl;
each R 7 Independently optionally substituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl, halogen, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted C 3 -C 10 Cycloalkyl C 1 -C 6 Alkyl, optionally substituted 5-to 14-membered heteroaryl, optionally substituted 4-to 14-membered heterocyclyl, -N (R) 7A ) 2 OR-OR 7A Wherein each R is 7A Independently H, optionally substituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted C 6 -C 10 Aryl, optionally substituted 5-to 10-membered heteroaryl or optionally substituted 4-to10-membered heterocyclyl, or two geminal R 7A The groups are combined with the attached atoms to form an optionally substituted 5-to 10-membered heteroaryl or an optionally substituted 4-to 10-membered heterocyclyl; or two twin R 7 The groups are combined with the attached atoms to form carbonyl groups;
R 8 is hydrogen, halogen, optionally substituted C 1 -C 6 Alkyl or optionally substituted C 3 -C 10 Cycloalkyl; and is also provided with
R 9 Hydrogen or halogen;
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound or pharmaceutically acceptable salt thereof has the structure of any one of compounds 1-308 in table 1A. In some embodiments, the compound or pharmaceutically acceptable salt thereof has the structure of any one of compounds 309-856 in table 1B.
Other embodiments and exemplary methods for synthetically producing these compounds are described herein.
Pharmaceutical use
The compounds described herein are useful in the methods of the invention and, while not being bound by theory, are believed to exert their ability to modulate the level, state and/or activity of BAF complexes, i.e., by inhibiting the activity of BRG1 and/or BRM proteins within BAF complexes in mammals. BAF complex-related disorders include, but are not limited to, BRG1 loss-of-function mutation-related disorders.
One aspect of the invention relates to methods of treating a disorder associated with a BRG1 loss-of-function mutation, such as cancer (e.g., non-small cell lung cancer, colorectal cancer, bladder cancer, unknown primary cancer, glioma, breast cancer, melanoma, non-melanoma skin cancer, endometrial cancer, or penile cancer) in a subject in need thereof. In some embodiments, the compound is administered in an amount and for a time effective to result in one or more (e.g., two or more, three or more, four or more) of: (a) reduced tumor size, (b) reduced tumor growth rate, (c) increased tumor cell death, (d) reduced tumor progression, (e) reduced number of metastases, (f) reduced metastasis rate, (g) reduced tumor recurrence, (h) increased subject survival, (i) increased subject progression-free survival.
Treatment of cancer may result in a decrease in the size or volume of the tumor. For example, after treatment, the tumor size is reduced by 5% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more) relative to its pre-treatment size. The size of the tumor can be measured by any reproducible measurement means. For example, the size of a tumor can be measured as the diameter of the tumor.
Treatment of cancer may further result in a reduction in the number of tumors. For example, after treatment, the number of tumors is reduced by 5% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more) relative to the number before treatment. The number of tumors can be measured by any reproducible measurement means, for example, the number of tumors can be measured by counting tumors that are visible to the naked eye or at a specified magnification (e.g., 2x, 3x, 4x, 5x, 10x, or 50 x).
Treatment of cancer may result in a reduction in the number of metastatic nodules in other tissues or organs distant from the primary tumor site. For example, after treatment, the number of metastatic nodules is reduced by 5% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more) relative to the number before treatment. The number of metastatic nodules can be measured by any reproducible means of measurement. For example, the number of metastatic nodules can be measured by counting metastatic nodules that are visible to the naked eye or at a specified magnification (e.g., 2x, 10x, or 50 x).
Treating cancer can result in an increase in the average survival time of a population of subjects treated according to the invention compared to an untreated population of subjects. For example, the average survival time increases by more than 30 days (more than 60 days, 90 days, or 120 days). The increase in average survival time of the population can be measured in any reproducible manner. The increase in average survival time of a population can be measured, for example, by calculating the average survival length of the population after starting treatment with a compound of the invention. The increase in average survival time of a population can also be measured, for example, by calculating the average survival length of the population after completion of a first round of treatment with a pharmaceutically acceptable salt of the invention.
Treating cancer may also result in reduced mortality in the treated population of subjects compared to the untreated population. For example, mortality is reduced by more than 2% (e.g., more than 5%, 10%, or 25%). The reduction in mortality of a population of subjects treated can be measured by any reproducible means, for example, by calculating the average number of disease-related deaths per unit time of the population after initiation of treatment with the pharmaceutically acceptable salts of the invention. The reduction in mortality of the population can also be measured, for example, by calculating the average number of disease-related deaths per unit time of the population after completion of the first round of treatment with the pharmaceutically acceptable salts of the invention.
Exemplary cancers that may be treated by the present invention include, but are not limited to, non-small cell lung cancer, colorectal cancer, bladder cancer, glioma, breast cancer, melanoma, non-melanoma skin cancer, endometrial cancer, esophageal gastric cancer, pancreatic cancer, hepatobiliary cancer (hepatobiliary cancer), soft tissue sarcoma, ovarian cancer, head and neck cancer, renal cell carcinoma, bone cancer, non-hodgkin's lymphoma, prostate cancer, embryonic tumor, germ cell tumor, cervical cancer, thyroid cancer, salivary gland cancer, gastrointestinal neuroendocrine tumor, uterine sarcoma, gastrointestinal stromal tumor, CNS cancer, thymus tumor, adrenal cortex cancer, appendiceal cancer, small intestine cancer, and penile cancer.
Combination preparation and use thereof
The compounds of the invention may be combined with one or more therapeutic agents. In particular, the therapeutic agent may be a therapeutic agent that treats or prophylactically treats any of the cancers described herein.
Combination therapy
The compounds of the invention may be used alone or in combination with additional therapeutic agents (e.g., other agents that treat cancer or symptoms associated therewith), or in combination with other types of therapies that treat cancer. In combination therapy, the dosage of one or more therapeutic compounds may be reduced from the standard dosage when administered alone. For example, the dosages may be determined empirically based on drug combinations and permutations, or may be derived by isoradiometric analysis (isobolographic analysis) (e.g., black et al, neurology 65: S3-S6, 2005). In this case, the dosage of the compounds should provide a therapeutic effect when combined.
In some embodiments, the second therapeutic agent is a chemotherapeutic agent (e.g., a cytotoxic agent or other chemical compound useful in treating cancer). These include alkylating agents, antimetabolites, folic acid analogs, pyrimidine analogs, purine analogs and related inhibitors, vinca alkaloids, epipodophyllotoxins, antibiotics, L-asparaginase, topoisomerase inhibitors, interferons, platinum coordination compounds, anthracenedione substituted ureas, methyl hydrazine derivatives, adrenocortical hormone inhibitors, adrenocortical hormone, progestins, estrogens, antiestrogens, androgens, antiandrogens and gonadotropin releasing hormone analogs. Also included are 5-fluorouracil (5-FU), leucovorin (LV), irinotecan, oxaliplatin, capecitabine, paclitaxel, and docetaxel. Non-limiting examples of chemotherapeutic agents include alkylating agents, such as thiotepa and cyclophosphamide; alkyl sulfonates such as busulfan, imperosulfan (endoprostufan) and piposulfan (piposulfan); aziridines, such as benzodopa (benzodopa), carboquinone (carboquone), methodolol (meturedapa) and Wu Ruiduo bar (uredapa); ethyleneimine and methyl melamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphamide and trimethylol melamine; acetogenins (especially bullatacin) and bullatacin (bullatacin)); camptothecins (including the synthetic analog topotecan); bryostatin (bryostatin); hua Gongsu (callystatin); CC-1065 (including adozelesin, carbozelesin, and bizelesin synthetic analogs thereof); cryptosporins (cryptosporins) (in particular cryptosporidium 1 and cryptosporidium 8); dolastatin (dolastatin); duocarmycin (including synthetic analogs, KW-2189 and CB1-TM 1); soft corallool (eleutherobin); a podocarpine (pancratistatin); a kind of creeping Sarcandylin (sarcandylin); spinostatin (spinostatin); nitrogen mustards, such as chlorambucil, napthalene, cyclophosphamide, estramustine, ifosfamide, mechlorethamine hydrochloride (mechlorethamine oxide hydrochloride), melphalan, noverichin, bennethol (phenaterine), prednisone, triamcinolone, uracil mustards; nitrosoureas such as carmustine, chloroureptin (chlorozotocin), fotemustine (fotemustine), lomustine (lomustine), nimustine (nimustine) and ranimustine (ranimustine); antibiotics, such as enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gamma 1 and calicheamicin omega 1 (see, e.g., agnew, chem. Intl. Ed England.:183-186 (1994)); dynomicins, including dynomicin A; bisphosphonates, such as chlorophosphonates; epothilones; and neocarcinomycin chromophores and related chromoproteins (chromoproteins) enediyne antibiotics chromophores), aclacinomycin (aclacinomycins), actinomycin (actinomycins), azaserine (azaserine), bleomycins (calicins), capsaicin (carmomycins), carmomycins (carminomycin), carminomycin (carminomycin), and neomycin (carminomycin), and norubicin (6-dactinomycin), and the derivatives of the antibiotics (leucins-6-daptomycin) (doxorubicin, including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolinodoxorubicin, and deoxydoxorubicin), epirubicin (epirubicin), escorubicin, idarubicin (idarubicin), doxycycline (marcelomicin), mitomycin (mitomycins) such as mitomycin C, mycophenolic acid (mycophenolic acid), norgamycin (nogalamycin), olivomycin (olivorins), pelomycin (peplomycin), pertumycin, puromycin, quelamycin, rodobiccin, streptomycin (strenigricin), streptomycin (streptomnigricin)(streptozocin), tuberculin (tubercidin), ubenimex, zinostatin, zorubicin; antimetabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogs such as, for example, dimethyl folic acid (denopterin), methotrexate, ptertrexate (pteroprerin), trimellite (trimellitate); purine analogs such as fludarabine, 6-mercaptopurine, thioxanthine (thiamipriline), thioguanine (thioguanine); pyrimidine analogs such as, for example, ambcitabine (ancitabine), azacytidine (azacitidine), 6-azauridine (6-azauridine), carmofur (carmofur), cytarabine, dideoxyuridine (dideoxyuridine), doxifluridine, enocitabine (enocitidine), fluorouridine; androgens, such as, for example, card Lu Gaotong (calasterone), drotasone propionate (dromostanolone propionate), epinastanol (epinastanol), melandrane (mepistane), testosterone (testolactone); an anti-adrenergic agent, such as aminoglutethimide (mitotane), trilostane (trilostane); folic acid supplements such as folinic acid (folinic acid); asilaone (aceglatone); aldehyde phosphoramidate glycoside (aldophosphamide glycoside); aminolevulinic acid (aminolevulinic acid); enuracil (eniluracil); amsacrine (amacrine); bei Qubu western (bestrebicil); bisantrene (bisantrene); edatraxate (edatraxate); difamin (defofamine); colchicine (demecolcine); deaquinone (diaziquone); eformitine (elfomithin); irinotecan acetate (elliptinium acetate); epothilones; etodolac (etoglucid); gallium nitrate; hydroxyurea; lentinan (lentinan); lonidamine (lonidamine); maytansinoids (maytansinoids) such as maytansine and ansamitocins (ansamitocins); mitoguazone (mitoguazone); mitoxantrone; mo Pi dipyridamole (mopidanmol); nitroamino acid (nitrone); penstatin (penstatin); phenamei (phenamit); pirarubicin (pirarubicin); losoxantrone (losoxantrone); podophylloic acid (podophyllinic acid); 2-ethyl hydrazide; procarbazine (procarbazine); / >Polysaccharide complexes (JHS Natural Products, eugene, or)eg.); raschig (razoxane); rhizomycin (rhizoxin); benzofurans (sizofurans); spirogermanium (spirogermanium); tenidamic acid (tenuazonic acid); triiminoquinone (triaziquone); 2,2',2 "-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verakurine A (verracurin A), cyclosporin a (roridin a) and An Gelie (anguidine)); uratam (urethan); vindesine (vindeline); dacarbazine (dacarbazine); mannomustine (mannomustine); dibromomannitol (mitobronitol); dibromodulcitol (mitolactol); pipobromine (pipobroman); ganciclovir (gacytosine); arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxanes, e.g.>Paclitaxel (Bristol-Myers Squibb Oncology, prencton, N.J.),>paclitaxel nanoparticle formulation without cremophor (cremophor-free), albumin modification (American Pharmaceutical Partners, schaumberg, ill.) and +.>Docetaxel (Rhone-Poulenc Rorer, antonny, france); chlorobutylamine (chloranbucil); />Gemcitabine; 6-thioguanine (6-thioguanine); mercaptopurine (mercaptopurine); methotrexate; platinum coordination compounds such as cisplatin, oxaliplatin, and carboplatin; vinblastine; plating; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; / >Vinorelbine; norxiaoling (novantrone); teniposide; eda traxas; daunomycin (daunomycin); aminopterin (aminopterin); hilded (xeloda); ibandronate (ibandronate); irinotecan (e.g., CPT-11); topoisomerase enzymesInhibitor RFS 2000; difluoromethyl ornithine (DMFO); retinoids, such as retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Suitable dosing regimens for combination chemotherapy, two or more chemotherapeutic agents may be used in a mixture for administration in combination with a first therapeutic agent as described herein, are known in the art and are described, for example, in Saltz et al (1999) Proc ASCO 18:233a and Douillard et al (2000) Lancet 355:1041-7.
In some embodiments, the second therapeutic agent is a therapeutic agent that is a biological agent such as a cytokine (e.g., an interferon or interleukin (e.g., IL-2)) for the treatment of cancer. In some embodiments, the biologic is an anti-angiogenic agent, e.g., an anti-VEGF agent, e.g., bevacizumabIn some embodiments, the biological agent is an immunoglobulin-based biological agent, such as a monoclonal antibody (e.g., a humanized antibody, a fully human antibody, an Fc fusion protein, or a functional fragment thereof), that agonizes a target to stimulate an anti-cancer response or antagonize an antigen important for cancer. Such agents include Rituxan (rituximab); zenapax (Daclizumab/Daclizumab); simulinet (Basiliximab); synagis (Palivizumab/Palivizumab); remicade (Infliximab/Infliximab); herceptin (Trastuzumab); mylotarg (Gituzumab ozymer/Gemtuzumab ozogamicin); campath (Alemtuzumab/Alemtuzumab); zevalin (temozolomide/Ibritumomab tiuxetan); HUMIRA (Adalimumab/Adalimiumab); xolair (Omalizumab/Omalizumab); bexxar (Tositumomab-L-131/Tositumomab-I-131); raptiva (efacient/Efalizumab); erbitux (Cetuximab/Cetuximab); avastin (Bevacizumab); tysabri (Natalizumab/Natalizumab); actmura (tolizumab/Tocilizumab); vicatib (Panitumumab); lucentis (Ranibizumab); soliris (Exkulizumab/Eculizumab); cimzia (cetuzumab/Certolizumab pegol); simmoni (Golimumab/Golimumab); LLARIS (Canokinumab); s is S telara (Uteteterumab/Utekinumab); arzerra (Ofatumumab/Ofatumumab); prolia (denomab); numax (mevaluzumab/Motavizumab); ABThrax (Lei Xiku mab/Raxibacumab); benlysta (Belimumab/Belimumab); yervoy (Ipilimumab/Ipilimumab); adcetris (Bentuximab/Brentuximab Vedotin); perjeta (Pertuzumab/Pertuzumab); kadcyla (ado-trastuzumab emtansine); and Gazyva (atozumab/Obinutuzumab). Antibody-drug conjugates are also included.
The second agent may be a therapeutic agent that is not a drug treatment. For example, the second therapeutic agent is radiation therapy, cryotherapy, hyperthermia, and/or surgical excision of tumor tissue.
The second agent may be a checkpoint inhibitor. In one embodiment, the checkpoint inhibitor is an inhibitory antibody (e.g., a monospecific antibody, e.g., a monoclonal antibody). Antibodies may be, for example, humanized or fully human. In some embodiments, the checkpoint inhibitor is a fusion protein, such as an Fc receptor fusion protein. In some embodiments, the checkpoint inhibitor is an agent, such as an antibody, that interacts with a checkpoint protein. In some embodiments, the checkpoint inhibitor is an agent, such as an antibody, that interacts with a ligand of a checkpoint protein. In some embodiments, the inhibitor of the checkpoint is an inhibitor of CTLA-4 (e.g., an inhibitory antibody or small molecule inhibitor) (e.g., an anti-CTLA 4 antibody, such as ipilimumab/Yervoy or tremelimumab). In some embodiments, the inhibitor of the checkpoint is an inhibitor of PD-1 (e.g., an inhibitory antibody or small molecule inhibitor) (e.g., nivolumab- Pembrolizumab/->Pi Deli bead mab/CT-011). In some embodiments, the checkpoint inhibitor is an inhibitor of PDL1 (e.g., an inhibitory antibody or small molecule inhibitor) (e.g., MPDL3280A/RG7446, MEDI4736, MSB0010718C, BMS 936559). In some embodiments, the checkpoint inhibitor is PDInhibitors of L2 (e.g., PDL2/Ig fusion proteins such as AMP) (e.g., inhibitory antibodies or Fc fusion or small molecule inhibitors). In some embodiments, the inhibitor of the checkpoint is an inhibitor of B7-H3 (e.g., MGA 271), B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK1, CHK2, A2aR, B-7 family ligand, or a combination thereof (e.g., an inhibitory antibody or small molecule inhibitor).
In any of the combination embodiments described herein, the first and second therapeutic agents are administered simultaneously or sequentially in either order. The first therapeutic agent may be administered immediately before or after the second therapeutic agent for up to 1 hour, up to 2 hours, up to 3 hours, up to 4 hours, up to 5 hours, up to 6 hours, up to 7 hours, up to 8 hours, up to 9 hours, up to 10 hours, up to 11 hours, up to 12 hours, up to 13 hours, 14 hours, up to 16 hours, up to 17 hours, up to 18 hours, up to 19 hours, up to 20 hours, up to 21 hours, up to 22 hours, up to 23 hours, up to 24 hours, or up to 1-7, 1-14, 1-21, or 1-30 days.
Pharmaceutical composition
The compounds of the invention are preferably formulated as pharmaceutical compositions for administration to mammals, preferably humans, in a biocompatible form suitable for in vivo administration. Accordingly, in one aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention in admixture with a suitable diluent, carrier or excipient.
The compounds of the present invention may be used in the form of the free base, in the form of salts, solvates and as prodrugs. All forms are within the scope of the invention. According to the methods of the present invention, the compounds, or salts, solvates, or prodrugs thereof, may be administered to a patient in a variety of forms depending on the route of administration selected, as will be appreciated by those skilled in the art. The compounds of the invention may be administered, for example, by oral, parenteral, buccal, sublingual, nasal, rectal, patch, pump or transdermal administration, and the pharmaceutical compositions formulated accordingly. Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transdermal, nasal, intrapulmonary, intrathecal, rectal and topical modes of administration. Parenteral administration may be by continuous infusion over a selected period of time.
The compounds of the invention may be administered orally, for example with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard or soft shell gelatin capsules, or they may be compressed into tablets, or they may be incorporated directly into the diet of the diet. For oral therapeutic administration, the compounds of the present invention may be mixed with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups and wafers. The compounds of the invention may also be administered parenterally. Solutions of the compounds of the present invention may be prepared in water suitably mixed with a surfactant. Under normal conditions of storage and use, these formulations may contain a preservative to prevent microbial growth. Conventional methods and ingredients for selecting and preparing suitable formulations are described, for example, in Remington's Pharmaceutical Sciences (2003, 20 th edition) and The United States Pharmacopeia: the National Formulary (USP 24NF 19) published 1999. Pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid so that it can be easily administered by syringe. Compositions for nasal administration may be conveniently formulated as aerosols, drops, gels and powders. Aerosol formulations typically comprise a solution or fine suspension of the active substance in a physiologically acceptable aqueous or non-aqueous solvent and are typically presented in sterile form in single or multiple doses in sealed containers which may take the form of cartridges or refills for use with an aerosolization device. Alternatively, the sealed container may be a unitary dispensing device, such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve, which is intended to be discarded after use. When the dosage form comprises an aerosol dispenser, it will contain a propellant, which may be a compressed gas, such as compressed air or an organic propellant. Aerosol dosage forms may also take the form of pump-nebulizers. Compositions suitable for buccal or sublingual administration include tablets, troches and lozenges (pastilles), wherein the active ingredient is formulated with a carrier. The compositions for rectal administration are conveniently in the form of suppositories that contain conventional suppository bases. The compounds described herein may be administered intratumorally, for example as an intratumoral injection. Intratumoral injection is direct injection into the tumor vasculature and is particularly contemplated for discrete, solid, accessible tumors. Local, regional or systemic administration may also be suitable. The compounds described herein may advantageously be contacted by administering an injection or injections (e.g., at about 1cm intervals) to the tumor. In the case of surgical interventions, the invention may be used prior to surgery, for example to subject non-surgical tumours to resection. Continuous administration may also be applied where appropriate, for example by implantation of a catheter into a tumor or tumor vasculature.
The compounds of the invention may be administered to animals, e.g., humans, alone or in combination with a pharmaceutically acceptable carrier as described herein, the ratio of which is determined by the solubility and chemical nature of the compound, the route of administration selected, and standard pharmaceutical practice.
Dosage of
The dosage of the compounds of the invention and/or compositions comprising the compounds of the invention may vary depending on a number of factors, such as the pharmacodynamic properties of the compounds; the mode of administration; age, health, and weight of the recipient; the nature and extent of the symptoms; the frequency of treatment and the type of concurrent treatment (if any); and clearance of the compound in the animal to be treated. One skilled in the art can determine the appropriate dosage based on the factors described above. The compounds of the invention may be administered initially in a suitable dose, which may be adjusted as required for the clinical response. In general, satisfactory results are obtained when the compounds of the invention are administered to a human in daily doses of, for example, 0.05mg to 3000 mg. Dosage ranges include, for example, 10-1000mg.
Alternatively, the weight of the patient may be used to calculate the dose. For example, the dosage of the compound or pharmaceutical composition thereof administered to the patient may be from 0.1 to 100mg/kg.
Examples
The definitions for the following schemes and elsewhere herein are:
MeCN or ACN acetonitrile
AIBN azo bis-isobutyronitrile
Boc t-Butoxycarbonyl group
t-BuOK potassium tert-butoxide
DAST diethylaminosulfur trifluoride
DCE dichloroethane
DCM dichloromethane
DCPP-2HBF 4 1, 3-bis (dicyclohexylphosphino) propane bis (tetrafluoroborate)
DEA N, N-diethylamine
DMP Dess-Martin reagent (Dess-Martin periodinane), 1-tris
(Acetyloxy) -1, 1-dihydro-1, 2-
DIAD diisopropyl azodicarboxylate
DIBAL-H diisopropylaluminum hydride
DIEA or DIPEA N, N-diisopropylethylamine
DMA dimethylacetamide
DMAP 4- (dimethylamino) pyridine
DME 1, 2-dimethoxyethane
DMF N, N-dimethylamide
DMSO dimethyl sulfoxide
dppf bis (diphenylphosphino) ferrocene
EDCl 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride
ESI electrospray ionization
Et 3 N or TEA triethylamine
EA ethyl acetate
EtOH ethanol
FA formic acid
FCC flash column chromatography
HATU 2- (3H- [1,2,3] triazolo [4,5-b ] pyridin-3-yl) -l,1, 3-tetramethylisourea salt
HCl hydrochloric acid
HOAc acetic acid
HOBt hydroxybenzotriazole
HPLC high performance liquid chromatography
IPA isopropyl alcohol
LCMS liquid chromatography/mass spectrometry
m-CPBA 3-chloroperoxybenzoic acid
MeCN acetonitrile
MeI iodomethane
MeOH methanol
mL of
mmol millimoles
mg
MHz megahertz (MHz)
MS mass spectrometry
MTBE methyl tert-butyl ether
m/z mass/charge ratio
NBS N-bromosuccinimide
NIS N-iodosuccinimide
nm nanometer
NMR nuclear magnetic resonance
PE Petroleum ether
PhMe toluene
ppm parts per million
rt room temperature
RT retention time
SFC supercritical fluid chromatography
Spos Pd G3 (2-dicyclohexylphosphino-2 ',6' -dimethoxybiphenyl) [2- (2 ' -ammonia)
1,1' -biphenyl) palladium (II) mesylate
TBS t-Butyldimethylsilyl group
TBSCl t-butyldimethylsilyl chloride
TBDMS tertiary butyl dimethyl silyl chloride
TFA trifluoroacetic acid
TFAA trifluoroacetic anhydride
THF tetrahydrofuran
TMSCN trimethylsilyl cyanide
TosMIC tosylmethyl isocyanide
Zinc Ziram dimethyl dithiocarbamic acid
Material
All materials were obtained from commercial suppliers and used without further purification unless otherwise indicated. All reactions involving air or moisture sensitive reagents were carried out under nitrogen atmosphere.
Table 1C lists the compounds of the present invention prepared using the methods described herein.
TABLE 1C Compounds of the invention
/>
/>
/>
/>
Material
All materials were obtained from commercial suppliers and used without further purification unless otherwise indicated. All reactions involving air or moisture sensitive reagents were carried out under nitrogen atmosphere.
Example 1 preparation of intermediates
Intermediate 1.2,3-dihydro-5H-benzo [ e ] [1,4] oxathiepine-8-carboxylic acid 1, 1-dioxide
Step 1 preparation of methyl 4-bromo-2-mercaptobenzoate
To a solution of methyl 4-bromo-2-fluoro-benzoate (100 g,429.12 mmol) in DMF (1L) was added sodium sulfide (33.49 g,429.1mmol,18.0 mL) and the mixture was stirred at 30℃for 16h. The mixture was poured into water (6000 mL) and then the pH was adjusted to-3 with 2N HCl. The mixture was extracted with MTBE (3000 mL x 2). The combined organic phases were washed with brine (3000 ml x 3), dried over anhydrous Na 2 SO 4 Drying, filtering and concentrating under vacuum to obtain methyl 4-bromo-2-mercaptobenzoate (103 g, crude)As a yellow oil, which was used in the next step without further purification.
1 H NMR(400MHz,DMSO_d6)δ=7.91(d,J=1.6Hz,1H),7.83-7.81(m,1H),7.43-7.40(m,1H),5.58(br s,1H),3.83(s,3H)ppm。
Step 2 preparation of (4-bromo-2-mercaptophenyl) methanol
At 0 ℃ at N 2 To a mixture of methyl 4-bromo-2-mercaptobenzoate (103 g,416.82 mmol) in THF (1000 mL) was added LiAlH under an atmosphere 4 (15.82 g,416.82 mmol). The mixture was stirred at 0deg.C for 1hr. The mixture was poured into 1N HCl (2000 mL) and extracted with EtOAc (2000 mL. Times.2). The combined organic phases were washed with brine (2000 mL) and dried over anhydrous Na 2 SO 4 Drying, filtration and concentration in vacuo afforded (4-bromo-2-mercaptophenyl) methanol (88 g, crude) as a yellow oil which was used in the next step without further purification. 1 H NMR(400MHz,DMSO_d6)δ=7.59(s,1H),7.32(d,J=1.2Hz,2H),5.56-5.36(m,2H),4.39(s,2H)ppm。
Step 3 preparation of (4-bromo-2- (vinylthio) phenyl) methanol and (4-bromo-2- ((2-bromoethyl) thio) phenyl) methanol
To a mixture of (4-bromo-2-mercaptophenyl) methanol (85 g,387.95 mmol) in DMF (1700 mL) was added K 2 CO 3 (160.9 g,1.16 mol) and 1, 2-dibromoethane (218.6 g,1.16mol,87.8 mL), and the mixture was stirred at 25℃for 1hr. The mixture was then stirred at 70℃for a further 24h. The reaction mixture was poured into saturated NH 4 Cl (10L) was extracted with EA (3000 mL. Times.2). The combined organic layers were washed with brine (4000 mL x 2), with Na 2 SO 4 Drying, filtering and evaporating the filtrate to dryness. The residue was purified by silica gel column chromatography (PE/ea=50/1-5/1). Fractions were concentrated in vacuo to give (4-bromo-2- (vinylthio) phenyl) methanol (33.5 g,136.66mmol,35% yield) and (4-bromo-2- ((2-bromoethyl) thio) phenyl) methanol (10 g,30.67mmol,8% yield) as yellow oil.
(4-bromo-2- (vinylthio) phenyl) methanol: 1 H NMR(400MHz,CDCl 3 )δ=7.55(s,1H),7.45(d,J=2Hz,1H),7.43(d,J=2Hz,1H),6.49-6.42(m,1H),5.45(d,J=9.6Hz,1H),5.32(d,J=10.4Hz,1H),4.73(s,2H)ppm;
(4-bromo-2- ((2-bromoethyl) thio) phenyl) methanol: 1 H NMR(400MHz,CDCl 3 )δ=7.46(s,1H),7.33(d,J=2Hz,1H),7.09(d,J=2Hz,1H),6.67(s,2H),3.41-3.38(m,2H),3.25-3.23(m,1H)ppm。
step 4 preparation of (4-bromo-2- (vinylsulfonyl) phenyl) methanol
To (4-bromo-2- (vinylthio) phenyl) methanol (35.5 g,144.82 mmol) in MeOH (350 mL) and H 2 To the mixture in O (350 mL)(133.54 g,217.23 mmol) and the mixture was stirred at 25℃for 2h. Water (1500 mL) was added and the mixture extracted with EtOAc (1500 mL x 2). The combined organic phases were washed with brine (1000 mL x 2), dried over anhydrous Na 2 SO 4 Drying, filtration and concentration in vacuo afforded (4-bromo-2-vinylsulfonyl-phenyl) methanol (38.5 g, crude) as a yellow solid, which was used in the next step without further purification.
1 H NMR(400MHz,DMSO_d6)δ=7.98-7.95(m,2H),7.77-7.75(m,1H),7.22-7.15(m,1H),6.43-6.39(m,1H),6.31(d,J=10.0Hz,1H),5.62-5.59(m,1H),4.75(d,J=5.2Hz,2H)ppm。
Step 5 preparation of 8-bromo-2, 3-dihydro-5H-benzo [ e ] [1,4] oxathiepine 1, 1-dioxide
At 0 ℃ at N 2 To a mixture of (4-bromo-2-vinylsulfonyl-phenyl) methanol (38.5 g,138.9 mmol) in DMF (1000 mL) was added NaH (11.11 g,277.84mmol,60% purity) under an atmosphere. The mixture was stirred at 0deg.C for 1hr. The reaction mixture was poured into saturated NH 4 Cl (2L) was extracted with EA (2000 mL. Times.2). The combined organic phases were washed with brine (2000 mL) and dried over anhydrous Na 2 SO 4 Drying, filtering and concentrating in vacuum. Purification of the residue by column chromatography (SiO 2 PE: etOAc=50:1-5:1) concentrated in vacuo to give 8-bromo-2, 3-dihydro-5H-benzo [ e][1,4]Oxathiepine 1, 1-dioxide (26.5 g,95.62mmol,69% yield) as a white solid. 1 H NMR(400MHz,DMSO_d6)δ=7.99(d,J=2.0Hz,1H),7.92-7.90(m,1H),7.55(d,J=8.0Hz,1H),4.88(s,2H),4.20-4.17(m,2H),3.68-3.66(m,2H)ppm。
Step 6 preparation of 2, 3-dihydro-5H-benzo [ e ] [1,4] oxathiepine-8-carboxylic acid 1, 1-dioxide (intermediate 1)
To 8-bromo-2, 3-dihydro-5H-benzo [ e][1,4]Oxathiepine 1, 1-dioxide (8.8 g,31.75 mmol) in DMSO (90 mL) and H 2 1, 3-bis (dicyclohexylphosphino) propane bis (tetrafluoroborate) (3.89 g,6.35 mmol), K was added to a mixture in O (9 mL) 2 CO 3 (6.58 g,47.63 mmol) and Pd (OAc) 2 (712.90 mg,3.18 mmol). The mixture was purged 3 times with CO and then stirred at 100deg.C under CO atmosphere (15 psi) for 4h. Water (3000 mL) was added and the mixture extracted with EtOAc (500 mL. Times.2) and the organic phase was then discarded. The aqueous layer was adjusted to pH 3 with 1N HCl. The mixture was then extracted with EA (500 ml x 5). The combined organic phases were washed with brine (2000 mL) and dried over anhydrous Na 2 SO 4 Drying, filtering and concentrating in vacuum. The crude product was washed with MTBE (20 ml x 2), then filtered and the filter cake evaporated to dryness to give 2, 3-dihydro-5H-benzo [ e ]][1,4]Oxathiepine-8-carboxylic acid 1, 1-dioxide (15 g,61.92mmol,65% yield) as a white solid.
1 H NMR(400MHz,DMSO-d6)δ=8.43(s,1H),8.20-8.18(m,1H),7.72-7.70(m,1H),4.96(s,2H),4.23-4.20(m,2H),3.67-3.66(m,2H)ppm。
Intermediate 2.3,5-dihydro-2H- [1,4] oxathiepino [6,5-b ] pyridine-8-carboxylic acid 1, 1-dioxide
Step 1 preparation of 5-bromo-3-mercaptopicolinic acid methyl ester
To a solution of 5-bromo-3-fluoro-pyridine-2-carboxylic acid methyl ester (1 g,4.27 mmol) in DMF (10 mL) was added Na 2 S (333.49 mg,4.27 mmol). The mixture was stirred at 25℃for 2h. Three identical batches were combined and purified together. The mixture was diluted with water (50 mL) and adjusted to ph=5 with 1N aqueous HCl. The mixture was extracted with EA (50 ml x 2). The combined organic layers were washed with brine (50 mL x 2), dried over anhydrous Na 2 SO 4 Drying and concentrating to obtain 5-Methyl bromo-3-mercaptopicolinate (3.3 g, crude) as a brown oil. LCMS (ESI) M/z: [ M+H ]] + =247.8/249.8。
Step 2 preparation of (5-bromo-3-mercaptopyridin-2-yl) methanol
To a solution of methyl 5-bromo-3-mercaptopicolinate (3.3 g,13.30 mmol) in THF (33 mL) at 0deg.C was added LiAlH 4 (504.8 mg,13.30 mmol). The mixture was stirred at 25℃for 2h. The mixture was diluted with water (100 mL) and adjusted to ph=6 with 1N aqueous HCl. The mixture was then extracted with EA (100 ml x 2). By anhydrous Na 2 SO 4 The combined organic layers were dried and concentrated to give (5-bromo-3-mercaptopyridin-2-yl) methanol (1.66 g,7.54 mmol) as a brown oil.
LCMS(ESI)m/z:[M+H] + =219.8/221.8。
Step 3 preparation of (5-bromo-3- (vinylthio) pyridin-2-yl) methanol
To a solution of (5-bromo-3-mercaptopyridin-2-yl) methanol (1.66 g,7.54 mmol) in DMF (15 mL) was added K 2 CO 3 (3.13 g,22.63 mmol) and 1, 2-dibromoethane (7.08 g,37.71mmol,2.85 mL). The mixture was stirred at 60℃for 12h. The mixture was diluted with water (100 mL) and extracted with EA (100 mL x 2). By anhydrous Na 2 SO 4 The combined starch organic layers were dried and concentrated to give a residue. Purification of the residue by flash chromatography on silica gel20g/>Silica gel flash column, eluent of 0-100% ethyl acetate/petroleum ether). The eluate was concentrated to give (5-bromo-3- (vinylthio) pyridin-2-yl) methanol (600 mg,2.44mmol,32% yield) as a brown oil.
LCMS(ESI)m/z:[M+H] + =245.9/247.9;
1 HNMR(400MHz,DMSO-d 6 )δ=8.53(d,J=2.0Hz,1H),7.92(d,J=2.0Hz,1H),6.81-6.74(m,1H),5.64-5.51(m,2H),5.32-5.29(m,1H),4.54(d,J=6.0Hz,2H)ppm。
Step 4 preparation of (5-bromo-3- (vinylsulfinyl) pyridin-2-yl) methanol
To a solution of (5-bromo-3- (vinylthio) pyridin-2-yl) methanol (600 mg,2.44 mmol) in MeOH (6 mL) at 0deg.C was slowly added water (6 mL)(824.27 mg,1.34 mmol). The mixture was stirred at 25℃for 1hr. With saturated Na 2 SO 3 The mixture was quenched with aqueous (30 mL) and extracted with EA (30 mL. Times.2). By anhydrous Na 2 SO 4 The combined organic layers were dried and concentrated to give a residue. The residue was purified by flash chromatography on silica gel (+.>12gSilica gel flash column, eluent of 0-100% ethyl acetate/petroleum ether). The eluate was concentrated to give (5-bromo-3- (vinylsulfinyl) pyridin-2-yl) methanol (500 mg,1.91mmol,78.25% yield) as a colorless oil.
1 HNMR(400MHz,DMSO-d 6 )δ=8.74(d,J=2.4Hz,1H),8.17(d,J=2.0Hz,1H),7.18-7.12(m,1H),6.09-6.02(m,2H),5.95(d,J=9.6Hz,1H),4.85-4.78(m,1H),4.73-4.66(m,1H)ppm。
Step 5 preparation of 8-bromo-3, 5-dihydro-2H- [1,4] oxathiepino [6,5-b ] pyridine 1-oxide
To a solution of (5-bromo-3- (vinylsulfinyl) pyridin-2-yl) methanol (500 mg,1.91 mmol) in DMF (5 mL) was added NaH (152.59 mg,3.81mmol,60% purity) at 0deg.C. The mixture was stirred at 0℃for 2h. With saturated NH 4 The mixture was quenched with aqueous Cl (30 mL) and extracted with EA (30 mL x 2). With no Na 2 SO 4 The combined organic layers were dried with water and concentrated to give a residue. Purification of the residue by flash chromatography on silica gel 12g/>Silica gel flash column, eluent of 0-10% ethyl acetate/petroleum ether). Concentrating the eluate to obtain 8-bromo-3, 5-dihydro-2H- [1,4]]Oxathiepino [6,5-b ]]Pyridine 1-oxide (350 mg,1.34mmol,70% yield) was a colorless oil. />
1 HNMR(400MHz,DMSO-d 6 )δ=8.75(d,J=2.4Hz,1H),8.19(d,J=2.0Hz,1H),4.91-4.74(m,2H),4.43-4.34(m,1H),4.21-4.18(m,1H),3.65-3.56(m,1H),3.49-3.44(m,1H)ppm。
Step 6 preparation of 1-oxide of 3, 5-dihydro-2H- [1,4] oxathiepino [6,5-b ] pyridine-8-carboxylic acid
To 8-bromo-3, 5-dihydro-2H- [1,4]Oxathiepino [6,5-b ]]To a solution of pyridine 1-oxide (350 mg,1.34 mmol) in DMSO (4 mL) and water (120.27 mg,6.68mmol,120.27 uL) was added 1, 3-bis (dicyclohexylphosphino) propane bis (tetrafluoroborate) (81.75 mg, 133.52. Mu. Mol), K 2 CO 3 (276.82 mg,2.00 mmol) and Pd (OAc) 2 (29.98 mg, 133.52. Mu. Mol). The mixture was degassed and purged 3 times with CO. The mixture was stirred at 100deg.C under CO atmosphere (15 psi) for 12h. The mixture was filtered and washed with DMSO (2 mL) and water (2 mL). The filtrate was then adjusted to ph=6 with 1N aqueous HCl. The filtrate was purified by reverse phase HPLC (0.1% FA conditions). Concentrating the eluate to remove ACN, and lyophilizing to obtain 3, 5-dihydro-2H- [1,4]]Oxathiepino [6,5-b ]]Pyridine-8-carboxylic acid 1-oxide (70 mg,0.262mmol,20% yield) as a white solid.
LCMS(ESI)m/z:[M+H] + =227.9;
1 HNMR(400MHz,DMSO-d 6 )δ=9.03(d,J=2.0Hz,1H),8.51(d,J=2.0Hz,1H),5.01-4.88(m,2H),4.43-4.41(m,1H),4.18-4.15(m,1H),3.63-3.62(m,2H),3.52-3.48(m,2H)ppm。
Step 7 preparation of 3, 5-dihydro-2H- [1,4] oxathiepino [6,5-b ] pyridine-8-carboxylic acid 1, 1-dioxide (intermediate 2)
3, 5-dihydro-2H- [1,4] at 0deg.C]Oxathiepino [6,5-b ]]Pyridine-8-carboxylic acid 1-oxide (70 mg,0.309 mmol) in MeOH @0.7 mL) of the solution in water (0.7 mL)(284.07 mg, 462.07. Mu. Mol). The mixture was stirred at 25 ℃ for 1hr and the mixture was filtered. The filter cake was washed with MeOH (5 mL). Then using saturated Na 2 SO 3 And the solution quenched the filtrate. The solution was then purified by reverse phase HPLC (0.1% FA conditions). Concentrating the eluate to remove ACN, and lyophilizing to obtain 3, 5-dihydro-2H- [1,4]]Oxathiepino [6,5-b ]]Pyridine-8-carboxylic acid 1, 1-dioxide (36 mg, 136.16. Mu. Mol,44% yield) as a white solid. LCMS (ESI) M/z: [ M+H ]] + =243.9。
Intermediate 3.3,4-dihydro-2H-benzo [ b ] [1,4] oxathiepine-7-carboxylic acid 5, 5-dioxide
Step 1 preparation of 3-chlorosulfonyl 4-hydroxy-benzoic acid
To HSO 3 To a Cl solution (31 mL) was added 4-hydroxybenzoic acid (5.5 g,39.82 mmol) stepwise. The mixture was stirred at 20℃for 16h. Ice water (300 mL) was slowly added dropwise to the reaction mixture. The mixture was extracted with ethyl acetate (100 ml x 3). The combined organic layers were washed with brine (50 mL), and dried over Na 2 SO 4 Drying and vacuum concentration gave a residue. The crude product was triturated with PE (30 mL) at 20℃for 30min to give 3-chlorosulfonyl 4-hydroxy-benzoic acid (4.5 g,13.72mmol,74% yield) as a white solid. 1 HNMR(400MHz,DMSO-d 6 )δ=8.09-8.06(m,1H),7.82-7.75(m,1H),6.89-6.81(m,1H)ppm。
Step 2 preparation of 4-hydroxy-3-mercaptobenzoic acid
To a solution of 3-chlorosulfonyl-4-hydroxy-benzoic acid (1 g,4.23 mmol) in toluene (20 mL) was added PPh in portions 3 (3.88 g,14.79 mmol). The mixture was stirred at 90℃for 2h. The reaction was stopped by adding 10% NaOH solution (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The aqueous phase was adjusted to pH 2 with 1N HCl. With ethyl acetate(20 mL x 3) the mixture was extracted. With Na 2 SO 4 The combined organic layers were dried and concentrated in vacuo to give 4-hydroxy-3-mercaptobenzoic acid (0.62 g,3.64mmol,86.21% yield) as a yellow oil.
1 H NMR(400MHz,DMSO-d 6 )δ=12.34(s,1H),7.89-7.83(m,1H),7.61-7.52(m,1H),6.90-6.83(m,1H),5.01(s,1H)ppm。
Step 3 preparation of 4-hydroxy-3-mercaptobenzoic acid methyl ester
To a solution of 4-hydroxy-3-mercaptobenzoic acid (0.6 g,3.53 mmol) in MeOH (5 mL) was added dropwise H 2 SO 4 (352.84 mg,3.53mmol,191.76uL,98% purity). The mixture was stirred at 70℃for 40h. The reaction was stopped by the addition of water (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The combined organic layers were washed with brine (20 mL), and dried over Na 2 SO 4 Drying and concentration in vacuo afforded methyl 4-hydroxy-3-mercaptobenzoate (0.6 g, crude) as a white solid.
1 H NMR(400MHz,DMSO-d 6 )δ=11.30(s,1H),8.07-8.01(m,1H),7.77-7.71(m,1H),6.99-6.95(m,1H),3.80-3.78(m,3H)ppm。
Step 4 preparation of 3, 4-dihydro-2H-benzo [ b ] [1,4] oxathiepine-7-carboxylic acid methyl ester
To a solution of methyl 4-hydroxy-3-mercaptobenzoate (0.1 g, 542.85. Mu. Mol,1 eq) in DMF (5 mL) was added Cs 2 CO 3 (884.36 mg,2.71 mmol) 1, 3-dibromopropane (109.6 mg,0.543mmol,55 uL) was added dropwise. The mixture was stirred at 20℃for 2h. The reaction was stopped by the addition of water (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The combined organic layers were washed with brine (20 mL), and dried over Na 2 SO 4 Drying and vacuum concentration gave a residue. Purification of the residue by preparative TLC (SiO 2 Petroleum ether: ethyl acetate=1:1), and concentrating the eluate in vacuo to give 3, 4-dihydro-2H-benzo [ b ]][1,4]Oxathiepine-7-carboxylic acid methyl ester (65 mg,0.274mmol,51% yield) was a yellow oil.
LCMS(ESI)m/z:[M+H] + =225.1;
1 H NMR(400MHz,CDCl 3 )δ=8.09-8.02(m,1H),7.83-7.74(m,1H),7.03-6.94(m,1H),4.45-4.34(m,2H),3.93-3.84(m,3H),3.10-2.98(m,2H),2.34-2.22(m,2H)ppm。
Step 5 preparation of 3, 4-dihydro-2H-benzo [ b ] [1,4] oxathiepine-7-carboxylic acid methyl ester 5, 5-dioxide
To 3, 4-dihydro-2H-1, 5-benzoxathiepin-7-carboxylic acid methyl ester (60 mg, 267.53. Mu. Mol) in MeOH (5 mL) and H 2 To the mixture in O (5 mL)(493.40 mg, 802.58. Mu. Mol) and then the mixture was stirred at 20℃for 16h. By addition of saturated Na 2 SO 3 (30 mL) the reaction was quenched. The mixture was extracted with DCM (30 mL. Times.5). With Na 2 SO 4 The combined organic layers were dried and concentrated in vacuo to give 3, 4-dihydro-2H-benzo [ b ]][1,4]Oxathiepine-7-carboxylic acid methyl ester 5, 5-dioxide (66 mg,0.257mmol,96% yield) was a yellow oil.
1 H NMR(400MHz,CDCl3)δ=8.73-8.60(m,1H),8.32-8.16(m,1H),7.26-7.23(m,1H),4.43-4.30(m,2H),3.97-3.91(m,3H),3.48-3.36(m,2H),2.53-2.41(m,2H)ppm。
Step 6 preparation of 3, 4-dihydro-2H-benzo [ b ] [1,4] oxathiepine-7-carboxylic acid 5, 5-dioxide (intermediate 3)
To 3, 4-dihydro-2H-benzo [ b ]][1,4]Oxathiepine-7-carboxylic acid methyl ester 5, 5-dioxide (65 mg,0.254 mmol) in MeOH (3 mL) and H 2 NaOH (30.44 mg,0.761 mmol) was added gradually to the mixture in O (3 mL), and the mixture was stirred at 20deg.C for 2h. The reaction was concentrated in vacuo to give a residue. The residue was partitioned with EA (10 mL) and 1N NaOH solution (10 mL). The aqueous layer was adjusted to pH 1 with 1N HCl solution and extracted with EA (10 mL. Times.3). Concentrating the combined organic phases in vacuo to give 3, 4-dihydro-2H-benzo [ b ]][1,4]Oxathiepine-7-carboxylic acid 5, 5-dioxide (60 mg,0.248mmol,98% yield) as a yellow solid. LCMS (ESI) M/z: [ M+Na ]] + =265.2;
1 H NMR(400MHz,DMSO-d6)δ=8.42-8.30(m,1H),8.21-8.10(m,1H),7.41-7.29(m,1H),4.34-4.22(m,2H),2.31 -2.22(m,2H),1.81-1.71(m,2H)ppm。
Intermediate 4, 6-chloro-2, 3-dihydro-5H-benzo [ e ] [1,4] oxathiepine-8-carboxylic acid 1, 1-dioxide
Step 1 preparation of 4-bromo-2-chloro-6-fluorobenzoic acid methyl ester
To a solution of 4-bromo-2-chloro-6-fluorobenzoic acid (10 g,39.46 mmol) in MeOH (90 mL) was slowly added concentrated H 2 SO 4 (18.4 g,187.60mmol,10 mL) and then the mixture was stirred at 70deg.C for 8h. The mixture was concentrated in vacuo to remove part of the MeOH, then poured into saturated NaHCO 3 (200 mL) and then extracted with EA (200 mL. Times.2). The combined organic layers were washed with brine (100 mL. Times.2), and dried over Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave methyl 4-bromo-2-chloro-6-fluorobenzoate (9.2 g, crude) as a colorless oil which was used directly in the next step.
1 H NMR(400MHz,DMSO-d 6 )δ=7.87-7.76(m,2H),3.91(s,3H)ppm。
Step 2 preparation of 4-bromo-2-chloro-6-mercaptobenzoic acid methyl ester
To a solution of methyl 4-bromo-2-chloro-6-fluorobenzoate (7.2 g,26.92 mmol) in DMF (72 mL) was added Na 2 S (2.10 g,26.92 mmol) and then the mixture was stirred at 25℃for 2h. The mixture was diluted with water (300 mL) and the resulting mixture was acidified to pH 3 with 1N HCl solution and extracted with EA (200 mL x 2). The combined organic layers were washed with brine (250 ml x 2), and dried over Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave methyl 4-bromo-2-chloro-6-mercaptobenzoate (7 g, crude) as a yellow oil, which was used directly in the next step.
Step 3 preparation of (4-bromo-2-chloro-6-mercaptophenyl) methanol
To a mixture of methyl 4-bromo-2-chloro-6-mercaptobenzoate (9 g,31.97 mmol) in THF (90 mL) at 0deg.C was added LiAlH 4 (1.33 g,35.16 mmol) and then the mixture was stirred at 0deg.C for 1hr. The mixture was poured into HCl (1N, 200 mL) followed by EA (250 mL. Times.2)And (5) extracting. The combined organic layers were washed with brine (200 ml x 2), and dried over Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave (4-bromo-2-chloro-6-mercaptophenyl) methanol (5.8 g, crude) as a colorless oil.
Step 4 preparation of (4-bromo-2-chloro-6- (vinylthio) phenyl) methanol
To a mixture of (4-bromo-2-chloro-6-mercaptophenyl) methanol (5.7 g,22.48 mmol) in DMF (110 mL) was added K 2 CO 3 (9.32 g,67.44 mmol) and 1, 2-dibromoethane (21.12 g,112.41mmol,8.5 mL), and then the mixture was stirred at 25℃for 12h. The mixture was poured into water (200 mL) and extracted with EA (100 mL). With Na 2 SO 4 The combined organic layers were dried, filtered and concentrated in vacuo to give a residue. The residue was purified by column chromatography (PE/ea=10/1-1:1, sio 2 ) The eluate was evaporated to give (4-bromo-2-chloro-6- (vinylthio) phenyl) methanol (2.7 g,9.66mmol,43% yield) as a colorless oil.
1 H NMR(400MHz,DMSO-d 6 )δ=7.65(d,J=2.0Hz,1H),7.42(d,J=2.0Hz,1H),6.78-6.71(m,1H),5.61 -5.52(m,2H),5.25-5.23(m,1H),4.62(d,J=5.2Hz,2H)ppm。
Step 5 preparation of methyl 3-chloro-4- (hydroxymethyl) -5- (vinylthio) benzoate
To a mixture of (4-bromo-2-chloro-6- (vinylthio) phenyl) methanol (1000 mg,3.58 mmol) in MeOH (20 mL) and TEA (10 mL) was added Pd (OAc) 2 (80.30 mg, 357.68. Mu. Mol) and XPhos (3411 mg, 0.015 mmol) and then the mixture was degassed, purged 3 times with CO (15 psi) and then the mixture was stirred at 70℃under CO atmosphere (15 psi) for 8h. The mixture was diluted with water (20 mL), extracted with EA (15 mL x 3), the combined organic phases were washed with brine (30 mL) and Na 2 SO 4 Drying, filtering and vacuum concentration to obtain crude product. Purification of the residue by column chromatography (SiO 2 Petroleum ether/ethyl acetate=20/1-5/1). The fractions were concentrated in vacuo to give methyl 3-chloro-4- (hydroxymethyl) -5- (vinylthio) benzoate (650 mg,2.36mmol,66% yield) as a white solid.
1 H NMR(400MHz,DMSO-d 6 )δ=7.83(d,J=1.6Hz,1H),7.80(d,J=1.6Hz,1H),6.74-6.67(m,1H),5.62-5.51(m,2H),5.36-5.33(m,1H),4.70(d,J=5.2Hz,2H),3.87(s,3H)ppm。
Step 6 preparation of methyl 3-chloro-4- (hydroxymethyl) -5- (vinylsulfonyl) benzoate
To methyl 3-chloro-4- (hydroxymethyl) -5- (vinylthio) benzoate (500 mg,1.93 mmol) in H 2 To a mixture of O (5 mL) and MeOH (5 mL) was added(3.56 g,5.80 mmol) and the mixture was stirred at 25℃for 1hr. The mixture was diluted with water (200 mL) and extracted with EA (250 mL. Times.2) and saturated Na 2 SO 3 (150 mL. Times.2) and brine (100 mL) were washed with Na 2 SO 4 Drying, filtration, and concentration under reduced pressure gave methyl 3-chloro-4- (hydroxymethyl) -5-vinylsulfonyl-benzoate (560 mg, crude) as a yellow oil. LCMS (ESI) M/z: [ M+H ]] + =273.0; 1 H NMR(400MHz,DMSO-d 6 )δ=8.42(d,J=1.6Hz,1H),8.27(d,J=1.6Hz,1H),7.34-7.27(m,1H),6.47-6.31(m,2H),5.52-5.49(m,2H),4.98(d,J=5.2Hz,2H),3.91(s,3H)ppm。
Step 7 preparation of 6-chloro-2, 3-dihydro-5H-benzo [ e ] [1,4] oxathiepine-8-carboxylic acid 1, 1-dioxide (intermediate 4)
To a mixture of methyl 3-chloro-4- (hydroxymethyl) -5-vinylsulfonyl-benzoate (560 mg,1.93 mmol) in THF (18 mL) was added NaH (154.09 mg,3.85mmol,60% purity) at 0 ℃ and the mixture was stirred at 0 ℃ for 1hr. The mixture was diluted with water (10 mL) and MeOH (5 mL), then stirred at 25 ℃ for 15min, the resulting mixture diluted with water (100 mL), acidified to pH 2 with HCl (1N) and the resulting solution extracted with EA (150 mL x 2). The combined organic layers were washed with brine (200 mL), and dried over Na 2 SO 4 Drying, filtering, concentrating under reduced pressure to obtain 6-chloro-2, 3-dihydro-5H-benzo [ e ]][1,4]Oxathiepine-8-carboxylic acid 1, 1-dioxide (300 mg, crude) as a white solid.
1 H NMR(400MHz,DMSO-d 6 )δ=13.91-13.84(m,1H),8.38(d,J=1.6Hz,1H),8.22(d,J=1.6Hz,1H),5.19(s,2H),4.23-4.21(m,2H),3.79-3.77-3.74(m,2H)ppm。
Intermediate 5:6-fluoro-2, 3-dihydro-5H-benzo [ e ] [1,4] oxathiepine-8-carboxylic acid 1, 1-dioxide
Step 1 preparation of methyl 4-bromo-2-fluoro-6- ((4-methoxybenzyl) thio) benzoate
To a mixture of methyl 4-bromo-2, 6-difluoro-benzoate (5 g,19.92 mmol) and (4-methoxyphenyl) methyl mercaptan (3.07 g,19.92mmol,2.77 mL) in DMF (50 mL) was added Cs 2 CO 3 (12.98 g,39.84 mmol) and then the mixture was stirred at 60℃for 2h. The mixture was diluted with water (400 mL), extracted with EA (200 mL x 3), the combined organic layers were washed with brine (200 mL x 2), then Na 2 SO 4 Drying, filtration and concentration in vacuo afforded methyl 4-bromo-2-fluoro-6- ((4-methoxybenzyl) thio) benzoate (9 g, crude) as a yellow oil which was used directly in the next step.
1 H NMR(400MHz,DMSO-d 6 )δ=7.54-7.51(m,2H),7.29-7.26(m,2H),6.90-6.87(m,2H),4.29(s,2H),3.83(s,3H),3.73-3.72(m,3H)ppm。
Step 2 preparation of 4-bromo-2-fluoro-6-mercaptobenzoic acid methyl ester
A mixture of methyl 4-bromo-2-fluoro-6- ((4-methoxybenzyl) thio) benzoate (9 g,23.36 mmol) in TFA (138.60 g,1.22mol,90 mL) was stirred at 60℃for 2h. The mixture was evaporated, then saturated NaHCO 3 Neutralized to pH 7. The mixture was then extracted with EA (200 mL). The organic layer was separated with anhydrous Na 2 SO 4 And (5) drying. The organic phase was concentrated in vacuo to give methyl 4-bromo-2-fluoro-6-mercaptobenzoate (6 g, crude) as a yellow oil, which was used directly in the next step.
1 H NMR(400MHz,DMSO-d 6 )δ=7.77-7.61(m,2H),3.93(s,3H)ppm。
Step 3 preparation of (4-bromo-2-fluoro-6-mercaptophenyl) methanol
To 4-bromo-2-fluoro-6-mercaptobenzoic acid methyl ester (3.4 g,12.83 mmol) inTo a mixture in THF (34 mL) was added LiAlH 4 (535.5 mg,14.11 mmol) and then the mixture was stirred at 0deg.C for 1hr. The mixture was quenched with 1N HCl (100 mL) and extracted with EA (50 mL). The organic layer was separated with anhydrous Na 2 SO 4 Drying, filtration and concentration in vacuo afforded (4-bromo-2-fluoro-6-mercaptophenyl) methanol (3 g, crude) as a yellow oil, which was used directly in the next step.
1 H NMR(400MHz,DMSO-d 6 )δ=7.51(s,1H),7.32-7.24(m,1H),4.45(d,J=1.2Hz,2H)。
Step 4 preparation of (4-bromo-2-fluoro-6- (vinylthio) phenyl) methanol
To (4-bromo-2-fluoro-6-mercaptophenyl) methanol (3 g,12.65 mmol), K 2 CO 3 (5.25 g,37.96 mmol) in DMF (60 mL) was added 1, 2-dibromoethane (11.89 g,63.27 mmol) and the mixture was then stirred at 25℃for 15h. The mixture was poured into water (200 mL) and extracted with EA (100 mL). With Na 2 SO 4 The combined organic layers were dried, filtered and concentrated in vacuo to give a residue. The residue was purified by column chromatography (PE/ea=10/1, sio 2 ) The eluate was evaporated to give (4-bromo-2-fluoro-6- (vinylthio) phenyl) methanol (1.6 g,6.08mmol,48% yield) as a colorless oil.
1 H NMR(400MHz,DMSO-d 6 )δ=7.47-7.44(m,1H),7.30-7.29(m,1H),6.77-6.70(m,1H),5.59-5.52(m,2H),5.22-5.20(m,1H),4.51-4.46(m,2H)ppm。
Step 5 preparation of (4-bromo-2-fluoro-6- (vinylsulfinyl) phenyl) methanol
To a mixture of (4-bromo-2-fluoro-6- (vinylthio) phenyl) methanol (800 mg,3.04 mmol) in DCM (12 mL) was added m-CPBA (678.98 mg,3.34mmol,85% purity) at 0deg.C, and the mixture was stirred at 25deg.C for 1hr. By addition of saturated Na at 0deg.C 2 SO 3 The reaction mixture was quenched with aqueous solution (20 mL) and then with H 2 O (20 mL) was diluted and extracted with EA (100 mL. Times.3). The combined organic layers were washed with brine 100mL and with Na 2 SO 4 Drying, filtering, and concentrating under reduced pressure to obtain a residue. Purification of the residue by column chromatography (SiO 2 Petroleum ether/acetic acid ethyl esterEster=20/1-1/1). The fractions were concentrated in vacuo to give (4-bromo-2-fluoro-6- (vinylsulfinyl) phenyl) methanol (690 mg,2.47mmol,81% yield) as a yellow solid. LCMS (ESI) m/z: [ 79 BrM+H] + =278.9;
1 H NMR(400MHz,DMSO-d 6 )δ=7.74-7.71(m,1H),7.61-7.60(m,1H),7.12-7.06(m,1H),6.05-5.92(m,2H),5.85-5.82(m,1H),4.75-4.71(m,1H),4.63-4.58(m,1H)ppm。
Step 6 preparation of 8-bromo-6-fluoro-2, 3-dihydro-5H-benzo [ e ] [1,4] oxathiepine 1-oxide
To a mixture of (4-bromo-2-fluoro-6- (vinylsulfinyl) phenyl) methanol (650 mg,2.33 mmol) in DMF (40 mL) was added NaH (186.3 mg,4.66mmol,60% purity) at 0deg.C, and the mixture was stirred at 0deg.C for 1hr. Saturated NH with 50mL 4 The reaction solution was quenched with aqueous Cl and extracted with EA (50 ml x 3). The combined organic layers were washed with brine (60 mL. Times.3), and dried over Na 2 SO 4 Drying, filtering, and concentrating under reduced pressure to obtain a residue. Purification of the residue by column chromatography (SiO 2 Petroleum ether/ethyl acetate=10/1-1/1), concentrating the eluent under reduced pressure to obtain 8-bromo-6-fluoro-2, 3-dihydro-5H-benzo [ e ]][1,4]Oxathiepine 1-oxide (350 mg,1.25mmol,54% yield) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ=7.79(m,1H),7.38-7.35(m,1H),5.13(d,J=14.4Hz,1H),4.49-4.34(m,3H),3.47-3.41(m,1H),3.26-3.21(m,1H)ppm。
Step 7 preparation of 6-fluoro-2, 3-dihydro-5H-benzo [ e ] [1,4] oxathiepine-8-carboxylic acid 1-oxide
To 8-bromo-6-fluoro-2, 3-dihydro-5H-benzo [ e][1,4]Oxathiepine 1-oxide (320 mg,1.15 mmol), pd (OAc) 2 (12.87 mg, 57.32. Mu. Mol) and dicyclohexyl (3-dicyclohexylphosphonium propyl) phosphonium; ditetrafluoroborate (70.19 mg, 114.64. Mu. Mol) in DMSO (4 mL) and H 2 K was added to the mixture in O (0.2 mL) 2 CO 3 (475.33 mg,3.44 mmol) and then the mixture was stirred at 100deg.C under CO (15 psi) for 4h. The mixture was diluted with water (50 mL) and extracted with EA (30 mL x 3). The aqueous layer was acidified to ph=3 with HCl solution (2M) and concentrated with EA (100ml x 2) extraction. The combined organic phases were washed with brine, and dried over Na 2 SO 4 Drying, filtering and vacuum concentrating to obtain 6-fluoro-2, 3-dihydro-5H-benzo [ e ]][1,4]Oxathiepine-8-carboxylic acid 1-oxide (210 mg, crude) was a white solid, which was used directly in the next step. LCMS (ESI) M/z: [ M+H ] ] + =244.9。
Step 8 preparation of 6-fluoro-2, 3-dihydro-5H-benzo [ e ] [1,4] oxathiepine-8-carboxylic acid 1, 1-dioxide (intermediate 5)
To 6-fluoro-2, 3-dihydro-5H-benzo [ e][1,4]Oxathiepine-8-carboxylic acid 1-oxide (210.00 mg, 859.81. Mu. Mol) in MeOH (4 mL) and H 2 To the mixture in O (4 mL)(634.30 mg,1.03 mmol) and then the mixture was stirred at 25℃for 2h. The mixture was diluted with water (50 mL) and extracted with EA (30 mL x 3), the combined organic layers were washed with brine (40 mL x 2) and Na 2 SO 4 Drying, filtering and vacuum concentrating to obtain 6-fluoro-2, 3-dihydro-5H-benzo [ e ]][1,4]Oxathiepine-8-carboxylic acid 1, 1-dioxide (220 mg, crude) was a white solid, which was used directly in the next step.
Intermediate 6 and 7 (R) -2-methyl-2, 3-dihydro-5H-benzo [ e ] [1,4] oxathiepine-8-carboxylic acid 1, 1-dioxide and (S) -2-methyl-2, 3-dihydro-5H-benzo [ e ] [1,4] oxathiepine-8-carboxylic acid 1, 1-dioxide
Step 1 preparation of 8-bromo-2-methyl-3, 5-dihydro-2H-benzo [ e ] [1,4] oxathiepine 1, 1-dioxide
To 8-bromo-2, 3-dihydro-5H-benzo [ e ] at 0deg.C][1,4]To a solution of oxathiepine 1, 1-dioxide (380 mg,1.37mmol,641.03 ul) in DMF (5 mL) was added NaH (65.82 mg,1.65mmol,60% purity). The mixture was stirred at 0℃for 0.5h. MeI (233.55 mg,1.65mmol,102.43 uL) was then added slowly at 0deg.C. The mixture was stirred at 25℃for 1.5h. With saturated NH 4 The mixture was diluted with Cl solution (30 mL) and EtOAc (30 mL. Times.2) extraction. By anhydrous Na 2 SO 4 The combined starch organic layers were dried and concentrated to give a residue. The residue was purified by reverse phase HPLC (0.1% FA conditions). Concentrating the eluate to remove MeCN, and lyophilizing to obtain 8-bromo-2-methyl-3, 5-dihydro-2H-benzo [ e ]][1,4]Oxathiepine 1, 1-dioxide (100 mg, 309.11. Mu. Mol,23% yield) as a white solid.
LCMS(ESI)m/z:[M+H] + =291.0/292.9;
1 HNMR(400MHz,DMSO-d 6 )δ=7.99(d,J=2.0Hz,1H),7.95-7.92(m,1H),7.56(d,J=8.0Hz,1H),4.87(s,2H),4.27 -4.23(m,1H),4.00-3.95(m,1H),3.71-3.62(m,1H),1.14(d,J=7.2Hz,3H)ppm。
Step 2 preparation of 2-methyl-3, 5-dihydro-2H-benzo [ e ] [1,4] oxathiepine-8-carboxylic acid 1, 1-dioxide
To 8-bromo-2-methyl-3, 5-dihydro-2H-benzo [ e][1,4]Oxathiepine 1, 1-dioxide (100 mg, 343.45. Mu. Mol) in DMSO (1 mL) and H 2 1, 3-bis (dicyclohexylphosphino) propane bis (tetrafluoroborate) (21.03 mg, 34.35. Mu. Mol), K was added to a solution of O (30.95 mg,1.72mmol, 31. Mu.L) 2 CO 3 (71.20 mg, 515.18. Mu. Mol) and Pd (OAc) 2 (7.71 mg, 34.35. Mu. Mol). The flask was degassed and purged 3 times with CO. The mixture was stirred at 100deg.C under CO atmosphere (15 psi) for 4h. The mixture was filtered and washed with EA (2 mL) and water (2 mL). The mixture was then diluted with water (5 mL) and extracted with EA (5 mL. Times.2). The combined organic layers were discarded. The aqueous phase was adjusted to ph=6 with 1N aqueous HCl. The aqueous phase was then extracted with EA (5 ml x 2). The combined organic layers were washed with brine (5 ml x 2), dried over anhydrous Na 2 SO 4 Drying and concentrating to obtain 2-methyl-3, 5-dihydro-2H-benzo [ e ]][1,4]Oxathiepine-8-carboxylic acid 1, 1-dioxide (80 mg,0.290mol,85% yield) as a white solid.
LCMS(ESI)m/z:[M+H] + =256.9;
1 HNMR(400MHz,DMSO-d 6 )δ=8.44(d,J=1.6Hz,1H),8.22-8.19(m,1H),7.72(d,J=8.0Hz,1H),4.95(s,2H),4.29 -4.25(m,1H),4.03-3.98(m,1H),3.72-3.60(m,1H),1.14(d,J=6.8Hz,3H)ppm。
Step 3 preparation of (R) -2-methyl-3, 5-dihydro-2H-benzo [ e ] [1,4] oxathiepine-8-carboxylic acid 1, 1-dioxide (intermediate 6) and (S) -2-methyl-3, 5-dihydro-2H-benzo [ e ] [1,4] oxathiepine-8-carboxylic acid 1, 1-dioxide (intermediate 7)
By SFC (column: daicel ChiralPak IG (250X 30mm,10 um); mobile phase: [0.1% NH) 3 H 2 O MEOH]The method comprises the steps of carrying out a first treatment on the surface of the 30% -30% of B, 3.0;85 min) isolation of racemic 2-methyl-3, 5-dihydro-2H-benzo [ e ]][1,4]Oxathiepine-8-carboxylic acid 1, 1-dioxide. The eluate was concentrated to remove most of the solvent and adjusted to ph=6 with FA. The mixture was then extracted with DCM (20 ml x 2). By anhydrous Na 2 SO 4 The combined organic layers were dried and concentrated to give (R) -2-methyl-3, 5-dihydro-2H-benzo [ e ]][1,4]Oxathiepine-8-carboxylic acid 1, 1-dioxide (35 mg,0.136mmol,44% yield) as a white solid; and (S) -2-methyl-3, 5-dihydro-2H-benzo [ e][1,4]Oxathiepine-8-carboxylic acid 1, 1-dioxide (40 mg,0.156mmol,50.00% yield) as a white solid. Stereochemistry was arbitrarily assigned.
(R) -2-methyl-3, 5-dihydro-2H-benzo [ e ][1,4]Oxathiepine-8-carboxylic acid 1, 1-dioxide (intermediate 6): LCMS (ESI) M/z: [ M+Na ]] + =279.1;
Chiral SFC, IG-3_5CM_MEOH (DEA) _5_40_3ML_T35.M; rt= 1.729min;
(S) -2-methyl-3, 5-dihydro-2H-benzo [ e][1,4]Oxathiepine-8-carboxylic acid 1, 1-dioxide (intermediate 7): LCMS (ESI) M/z: [ M+Na ]] + =279.1;
Chiral SFC, IG-3_5CM_MEOH (DEA) _5_40_3ML_T35.M; rt= 1.897min.
Intermediate 8.6,7,8,9-tetrahydrothieno [3,2-b ] pyridine-3-carboxylic acid 5, 5-dioxide
Step 1 preparation of 2-bromo-5-chloro-pyridine-3-thiol
At 25 ℃ at N 2 To a mixture of 2-bromo-5-chloro-3-fluoro-pyridine (1.3 g,6.18mmol,1 eq) in DMF (20 mL) was added Na in one portion under an atmosphere 2 S (482.14 mg,6.18mmol, 259.22. Mu.L). The mixture was stirred at 25℃for 12h. The mixture was poured (100 mL). To the mixture was added aqueous HCl (2M) to adjust ph=3. The aqueous phase was extracted with ethyl acetate (50 ml x 2). The combined organic phases were washed with brine (50 mL x 1), dried over anhydrous Na 2 SO 4 Drying, filtration and concentration in vacuo afforded 2-bromo-5-chloro-pyridine-3-thiol (1.2 g,5.35mmol,86.52% yield) as a yellow solid.
LCMS(ESI)m/z:[ 79 BrM+H] + =225.8。
1 H NMR(400MHz,DMSO-d 6 )δ=8.33-8.23(m,1H),8.17-8.08(m,1H)ppm。
Step 2 preparation of 2-bromo-3- (but-3-en-1-ylsulfanyl) -5-chloropyridine
To a mixture of 2-bromo-5-chloro-pyridine-3-thiol (1.2 g,5.35 mmol) and but-3-en-1-ol (385.41 mg,5.35mmol,459.91 μl) in THF (10 mL) was added PPh 3 (2.10 g,8.02 mmol) followed by N at 0deg.C 2 DEAD (1.40 g,8.02mmol,1.46 mL) was added dropwise under an atmosphere. The mixture was stirred at 25℃for 12h. The mixture was poured into water (50 mL) and extracted with ethyl acetate (30 mL x 2). The combined organic phases were washed with brine (30 mL x 1), dried over anhydrous Na 2 SO 4 Drying, filtration and concentration in vacuo gave a residue. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=10/1). The eluate was concentrated to give 2-bromo-3- (but-3-en-1-ylsulfanyl) -5-chloropyridine (1.2 g,4.31mmol,81% yield) as a yellow oil. LCMS (ESI) m/z: [ 79 BrM+H] + =277.9。
1 H NMR(400MHz,CDCl 3 )δ=8.05-7.99(m,1H),7.34-7.28(m,1H),5.88-5.73(m,1H),5.16-5.01(m,2H),2.99-2.86(m,2H),2.48-2.35(m,2H)ppm。
Step 3 preparation of 3- (but-3-en-1-ylsulfanyl) -5-chloro-2-vinylpyridine
2-bromo-3- (but-3-en-1-ylsulfanyl) -5-chloropyridine (860 mg,3.09 mmol), potassium vinyltrifluoroborate (1.24 g,9.26 mmol), pd (dtbpf) Cl 2 (201.19 mg, 308.69. Mu. Mol) and K 3 PO 4 (1.97 g,9.26 mmol) in dioxane (12 mL) and H 2 Mixture in O (3 mL) at 80At a temperature of N 2 Stirring in the atmosphere for 1hr. Pouring the mixture into H 2 O (100 mL) was extracted with EA (30 mL. Times.3). The combined organic layers were washed with brine (20 mL x 2), dried over anhydrous Na 2 SO 4 Drying, filtering, and concentrating under reduced pressure to obtain a residue. The residue was purified by silica gel chromatography (PE-PE/ea=20/1). The eluate was concentrated under reduced pressure to give 3- (but-3-en-1-ylsulfanyl) -5-chloro-2-vinylpyridine (510 mg,2.26mmol, 73% yield) as a yellow oil. LCMS (ESI) M/z: [ M+H ] ] + =226.0。
1 H NMR(400MHz,CDCl 3 )δ=8.36(d,J=2.4Hz,1H),7.60(d,J=2.4Hz,1H),7.26-7.19(m,1H),6.41-6.36(m,1H),5.89-5.81(m,1H),5.57-5.53(m,1H),5.20-5.07(m,2H),2.97-2.93(m,2H),2.44-2.36(m,2H)ppm。
Step 4 preparation of 3-chloro-6, 7-dihydrothieno [3,2-b ] pyridine
3- (but-3-en-1-ylsulfanyl) -5-chloro-2-vinylpyridine (250 mg,1.11 mmol) and benzylidene- [1, 3-bis (2, 4, 6-trimethylphenyl) imidazolidin-2-ylidene]-dichloro-ruthenium; a mixture of tricyclohexylphosphine (Grubbs II) (94.0 mg,0.111 mol) in DCM (12 mL) was N at 25 ℃ 2 The atmosphere was stirred for 16 h. The solution was concentrated in vacuo. The residue was purified by silica gel chromatography (PE-PE/ea=20/1). Concentrating the eluent under reduced pressure to obtain 3-chloro-6, 7-dihydrothieno [3,2-b ]]Pyridine (110 mg, 556.44. Mu. Mol,50% yield) was a yellow oil. LCMS (ESI) M/z: [ M+H ]] + =198.0。
1 H NMR(400MHz,CDCl 3 )δ=8.40(d,J=2.0Hz,1H),7.71(d,J=1.6Hz,1H),6.81-6.71(m,1H),6.32-6.26(m,1H),3.10-3.05(m,2H),2.88-2.81(m,2H)。
Step 5 preparation of 6, 7-Dihydrothieno [3,2-b ] pyridine-3-carboxylic acid
3-chloro-6, 7-dihydrothieno [3,2-b]Pyridine (50 mg,0.253 mol), K 2 CO 3 (52.44mg,0.379mol)、Pd(OAc) 2 (2.84 mg, 12.65. Mu. Mol), 1, 3-bis (dicyclohexylphosphino) propane bis (tetrafluoroborate) (15.49 mg, 25.29. Mu. Mol) and H 2 A mixture of O (100. Mu.L) in DMSO (1 mL) was stirred at 100deg.C under CO atmosphere (15 psi) for 4h. Pouring the mixture into H 2 O (10 mL), with EA (10 mL)x 2) extraction. The organic phase was discarded. The aqueous phase was acidified to ph=3 with HCl (1M) and extracted with EA (10 ml x 3). The combined organic layers were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Drying, filtering, concentrating under reduced pressure to obtain 6, 7-dihydro-thieno [3,2-b ] ]Pyridine-3-carboxylic acid (28 mg, 135.10. Mu. Mol,53.4% yield) was a white solid. LCMS (ESI) M/z: [ M+H ]] + =207.9。
1 H NMR(400MHz,CDCl 3 )δ=8.36(d,J=2.4Hz,1H),7.60(d,J=2.4Hz,1H),7.26-7.19(m,1H),6.41-6.36(m,1H),5.89-5.81(m,1H),5.57-5.53(m,1H),5.20-5.07(m,2H),2.97-2.93(m,2H),2.44-2.36(m,2H)ppm。
Step 6 preparation of 6,7,8, 9-tetrahydrothieno [3,2-b ] pyridine-3-carboxylic acid
To 6, 7-dihydrothieno [3,2-b ] at 25 DEG C]To a mixture of pyridine-3-carboxylic acid (28 mg, 135.10. Mu. Mol) in MeOH (5 mL) was added Pd/C (wet, 50mg,10% purity). By H 2 The mixture was purged 3 times at 25℃under H 2 Stirring was carried out in an atmosphere (15 psi) for 30min. The mixture was filtered, and the filtrate was concentrated under reduced pressure to give 6,7,8, 9-tetrahydrothieno [3,2-b ]]Pyridine-3-carboxylic acid (23 mg, 109.91. Mu. Mol,81.2% yield) was a white solid. LCMS (ESI) M/z: [ M+H ]] + =210.0。
1 H NMR(400MHz,DMSO-d 6 )δ=8.81(d,J=2.0Hz,1H),8.20(d,J=2.0Hz,1H),3.23-3.17(m,2H),2.88-2.78(m,2H),2.11-1.96(m,2H),1.76-1.62(m,2H)ppm。
Step 7 preparation of 5, 5-dioxo-6, 7,8, 9-tetrahydrothieno [3,2-b ] pyridine-3-carboxylic acid (intermediate 8)
To 6,7,8, 9-tetrahydrothieno [3,2-b ] at 25 DEG C]Pyridine-3-carboxylic acid (23 mg, 109.91. Mu. Mol) in MeOH (1 mL) and H 2 To the mixture in O (1 mL)(67.57 mg, 109.9. Mu. Mol). The mixture was stirred at 25℃for 4h. With saturated Na 2 SO 3 (20 mL) the mixture was quenched, acidified to ph=2 with HCl (1M) and extracted with EA (20 mL x 2). The combined organic layers were washed with brine, dried over anhydrous Na 2 SO 4 Drying, filtering, and depressurizingConcentrating to obtain 5, 5-dioxo-6, 7,8, 9-tetrahydrothieno [3,2-b ]]Pyridine-3-carboxylic acid (18 mg, 74.61. Mu. Mol,68% yield) was a white solid.
LCMS(ESI)m/z:[M+H] + =241.9;
1 H NMR(400MHz,DMSO-d 6 )δ=9.14(d,J=2.0Hz,1H),8.55(d,J=2.1Hz,1H),3.53-3.51(m,2H),3.17(br d,J=5.2Hz,2H),2.19-2.13(m,2H),1.82(br d,J=3.2Hz,2H)ppm。
Intermediate 9:4, 5-dihydro-2H-benzo [ d ] [1,3] oxathiepine-8-carboxylic acid 1, 1-dioxide
Step 1 preparation of (6-bromobenzo [ b ] thiophen-2-yl) boronic acid
At-70 ℃ at N 2 In the atmosphere to 6-bromobenzo [ b ]]To a mixture of thiophene (8 g,37.54 mmol) in THF (80 mL) was added LDA (2M, 22.53 mL) dropwise. The mixture was stirred at-70deg.C for 1hr. Triisopropyl borate (8.47 g,45.05mmol,10.36 mL) was then added to the mixture at-70℃and the mixture was stirred for 1hr. H is added to the mixture at-70 DEG C 2 SO 4 (7.36 g,75.08mmol,4.00 mL) and the mixture was stirred at 25℃for 1hr. The mixture was poured into water (300 mL) and extracted with ethyl acetate (200 mL x 2). The combined organic phases were washed with brine (200 ml x 1), dried over anhydrous Na 2 SO 4 Drying, filtering and concentrating in vacuum. The residue was triturated with PE/mtbe=10/1 (50 mL). The suspension was filtered. Drying the filter cake in a pump to give (6-bromobenzo [ b)]Thiophen-2-yl) boronic acid (7.3 g,28.41mmol,76% yield) was a pale yellow solid. 1 H NMR(400MHz,DMSO-d6)δ=8.58-8.53(m,2H),8.28-8.24(m,1H),7.96-7.93(m,1H),7.88-7.84(m,1H),7.54-7.46(m,1H)ppm。
Step 2 preparation of 6-bromobenzo [ b ] thiophen-2 (3H) -one
At 25 ℃ at N 2 In the atmosphere to (6-bromobenzo [ b ]]Thiophene-2-yl) boronic acid (6.5 g,25.30 mmol) in EtOH (78 mL) was added dropwise H 2 O 2 (38.35 g,338.24mmol,32.50 mL). The mixture was stirred at 25 ℃ Stirring for 1hr. The mixture was filtered. By H 2 O (50 mL) washing the filter cake, and vacuum drying to obtain 6-bromobenzo [ b ]]Thiophen-2 (3H) -one (4.2 g,18.33mmol,72% yield) was a brown solid. LCMS (ESI) M/z: [ M+H ]] + =214.8,216.9。
1 H NMR(400MHz,CDCl 3 )δ=7.53-7.47(m,1H),7.38-7.32(m,1H),7.16(d,J=8.0Hz,1H),4.06-3.84(m,2H)ppm。
Step 3 preparation of 2- (4-bromo-2-mercaptophenyl) ethan-1-ol
At 25 ℃ at N 2 In the atmosphere to 6-bromobenzo [ b ]]NaBH was added in portions to a mixture of thiophen-2 (3H) -one (4.2 g,18.33 mmol) in EtOH (67 mL) 4 (3.47 g,91.67 mmol). The mixture was stirred at 80℃for 30min. The mixture was cooled to 25 ℃. To this mixture was slowly added aqueous HCl (1M) to adjust ph=2. The mixture was poured into water (200 mL) and extracted with ethyl acetate (100 mL x 2). The combined organic phases were washed with brine (100 mL), with anhydrous Na 2 SO 4 Drying, filtering and concentrating in vacuum. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=2/1). The eluate was concentrated to give 2- (4-bromo-2-mercaptophenyl) ethan-1-ol (3.6 g,15.44mmol,84% yield) as a yellow oil. 1 H NMR(400MHz,DMSO-d6)δ=7.61(d,J=2.0Hz,1H),7.24-7.22(m,1H),7.13(d,J=8.2Hz,1H),5.58(s,1H),4.97-4.49(m,1H),3.59-3.57(m,2H),2.71-2.69(m,2H)。
Step 4 preparation of 8-bromo-4, 5-dihydrobenzo [ d ] [1,3] oxathiepine
At 0 ℃ at N 2 To a mixture of 2- (4-bromo-2-mercaptophenyl) ethan-1-ol (500 mg,2.14 mmol) in DMF (50 mL) was added NaH (257.37 mg,6.43 mmol) in portions under an atmosphere. The mixture was stirred at 25℃for 30min. Then at 0 ℃ under N 2 To the mixture was added dropwise chloromethane (416.13 mg,2.36mmol, 171. Mu.L) in DMF (1 mL) under an atmosphere. The mixture was stirred at 25℃for 1.5h. The mixture was poured into saturated NH 4 Cl (10 mL) was extracted with ethyl acetate (10 mLx 2). The combined organic phases were washed with brine (10 mL), with anhydrous Na 2 SO 4 Drying, filtering and concentrating in vacuum. Purification of the residue by silica gel chromatography (petroleum ether/acetic acidEthyl = 10/1). Concentrating the eluate to obtain 8-bromo-4, 5-dihydrobenzo [ d ]][1,3]Oxathiepine (50 mg,0.189mmol,9% yield) was a yellow oil.
LCMS(ESI)m/z:[M+H] + =246.2,248.0。
1 H NMR(400MHz,DMSO-d6)δ=7.67-7.60(m,1H),7.47-7.38(m,1H),7.31-7.22(m,1H),5.00-4.86(m,2H),3.81 -3.67(m,2H),3.12-3.09(m,2H)ppm。
Step 5 preparation of 4, 5-Dihydrobenzo [ d ] [1,3] oxathiepine-8-carboxylic acid
To 8-bromo-4, 5-dihydrobenzo [ d ]][1,3]Oxathiepine (50 mg, 203.97. Mu. Mol), pd (OAc) 2 (4.58 mg, 20.40. Mu. Mol), 1, 3-bis (dicyclohexylphosphino) propane bis (tetrafluoroborate) (24.98 mg, 40.79. Mu. Mol) and K 2 CO 3 (56.38 mg,0.408 mmol) in DMSO (2 mL) and H 2 The solution in O (0.2 mL) was degassed under vacuum and purged several times with CO. The mixture was stirred at 100deg.C under CO atmosphere (15 psi) for 2h. The mixture was poured into water (20 mL) and extracted with ethyl acetate (10 mL x 2). The organic layer was discarded. Aqueous HCl (1M) was added to the aqueous phase to adjust ph=3. The mixture was extracted with ethyl acetate (10 ml x 2). The combined organic phases were washed with brine (10 mL), with anhydrous Na 2 SO 4 Drying, filtering, vacuum concentrating to obtain 4, 5-dihydrobenzo [ d ]][1,3]Oxathiepine-8-carboxylic acid (40 mg, 190.25. Mu. Mol,93% yield) was a yellow solid. LCMS (ESI) M/z: [ M+H ]] + =211.1。
Step 6 preparation of 4, 5-dihydro-2H-benzo [ d ] [1,3] oxathiepine-8-carboxylic acid 1, 1-dioxide (intermediate 9)
At 25 ℃ at N 2 In the atmosphere to 4, 5-dihydrobenzo [ d ]][1,3]To a mixture of oxathiepine-8-carboxylic acid (20 mg, 95.13. Mu. Mol) in DCM (1 mL) was added mCPBA (48.28 mg, 237.81. Mu. Mol,85% purity) in portions. The mixture was stirred at 25℃for 12h. The mixture was filtered and the filtrate was concentrated. The residue was purified directly by reverse phase column (FA). The eluate was concentrated to remove MeCN. Lyophilizing the aqueous phase to obtain 4, 5-dihydrobenzo [ d ]][1,3]Oxathiepine-8-carboxylic acid 1, 1-dioxide (20 mg, 82.56. Mu. Mol,86.79% yield) as a white solid. LCMS (ESI) M/z: [ M+H ] 2 O] + =260.0;
1 H NMR(400MHz,DMSO-d6)δ=8.38(d,J=1.6Hz,1H),8.16-8.14(m,1H),7.62(d,J=7.8Hz,1H),4.99(s,2H),4.01-4.00(m,2H),3.42(s,2H)ppm。
Intermediate 10, 1-dioxide of 4, 5-dihydro-2H-benzo [ d ] [1,3] oxathiepine-8-carboxylic acid
Step 1 preparation of 4-bromo-2- [ (4-methoxyphenyl) methylsulfanyl ] benzonitrile
To a solution of 4-bromo-2-fluoro-benzonitrile (10 g,50.00 mmol) and (4-methoxyphenyl) methyl mercaptan (7.71 g,50.00 mmol) in DMF (100 mL) was added Cs 2 CO 3 (16.29 g,50.00 mmol) and the mixture was stirred at 60℃for 2h. The reaction mixture was poured into water (1000 mL), the solution extracted with EA (1000 mL. Times.3), the combined organic layers were washed with brine (500 mL), and dried over Na 2 SO 4 Drying, filtering and concentrating to obtain 4-bromo-2- [ (4-methoxyphenyl) methylsulfanyl]Benzonitrile (13 g, crude) as a white solid.
Step 2 preparation of [ 4-bromo-2- [ (4-methoxyphenyl) methylsulfanyl ] phenyl ] methylamine
At 0 ℃ at N 2 In the atmosphere to 4-bromo-2- [ (4-methoxyphenyl) methylsulfanyl]To a solution of benzonitrile (13 g,38.90 mmol) in THF (150 mL) was added LiAlH 4 (1.62 g,42.78 mmol) and the mixture was stirred at 0deg.C for 1hr. To the mixture was poured water (1.62 g) and 15% NaOH solution (2.5 mL), the solution was poured into EA (500 mL), the solution was filtered, and the filtrate was concentrated to give [ 4-bromo-2- [ (4-methoxyphenyl) methylsulfanyl]Phenyl group]Methylamine (13 g, crude) was a yellow oil.
1H NMR(400MHz,DMSO-d 6 )δ=7.48-7.47(m,1H),7.21-7.20(m,1H),7.19-7.18(m,3H),6.85-6.82(m,2H),4.08(s,2H),3.80-3.79(m,5H)ppm。
Step 3 preparation of [2- [ [2- (aminomethyl) -5-bromo-phenyl ] disulfanyl ] -4-bromo-phenyl ] methylamine
A mixture of [ 4-bromo-2- [ (4-methoxyphenyl) methylsulfanyl ] phenyl ] methylamine (13 g,38.43 mmol) in TFA (130 mL) was stirred at 60℃for 16h. The reaction mixture was concentrated to give a residue. The residue was purified by reverse phase HPLC (0.1% FA conditions). The solution was lyophilized to give aminomethyl) -5-bromo-phenyl ] disulfanyl ] -4-bromo-phenyl ] methylamine (3.5 g,7.20mmol,19% yield) as a white solid.
LCMS(ESI)m/z:[ 79 BrM+H] + =434.8。
1H NMR(400MHz,DMSO-d 6 )δ=8.35(br s,3H),7.55(s,2H),7.50-7.37(m,1H),4.05(s,2H)ppm。
Step 4 preparation of 8-bromo-4, 5-dihydro-1, 4-benzothiazepine (benzothiazepin) -3-one
To aminomethyl) -5-bromo-phenyl]Disulfanyl group]-4-bromo-phenyl]To a solution of methylamine (1 g,2.30 mmol) in THF (15 mL) was added NaBH 4 (261.37 mg,6.91 mmol) and the mixture was stirred at 30℃for 2h. TEA (11.52 mmol,1.60 mL), 2-chloroacetyl chloride (312.13 mg,2.76 mmol) was then added to the solution and the mixture was stirred at 30deg.C for 3h. The reaction mixture was poured into water (100 mL) and extracted with EA (100 mL x 3). The combined organic layers were washed with brine (200 mL), and dried over Na 2 SO 4 Drying, filtering and concentrating to obtain a residue. Purification of the residue by column chromatography (SiO 2 Petroleum ether/ethyl acetate=10:1-0:1), the solution was concentrated to give 8-bromo-4, 5-dihydro-1, 4-benzothiazepin-3-one (300 mg,871.29 μmol,38% yield) as a white solid.
LCMS(ESI)m/z:[ 79 BrM+H] + =260.0。
1H NMR(400MHz,DMSO-d 6 )δ=7.37(d,J=2.0Hz,1H),7.24–7.22(m,1H),7.07(d,J=8.0Hz,1H),4.45(s,2H),3.89(s,2H)ppm。
Step 5 preparation of 3-oxo-4, 5-dihydro-1, 4-benzothiazepine-8-carboxylic acid (intermediate 10)
To a solution of 8-bromo-4, 5-dihydro-1, 4-benzothiazepin-3-one (280 mg,1.08 mmol) in DMSO (5 mL) was added dicyclohexyl (3-dicyclohexylphosphonium propyl) phosphonium; ditetrafluoroborate (66.41 mg, 108.47. Mu. Mol), K 2 CO 3 (224.88mg,1.63mmol)、Pd(OAc) 2 (24.35mg,10847. Mu. Mol) and H 2 O (3.91 mg, 216.94. Mu. Mol) and the mixture was stirred in a CO atmosphere (15 psi) at 100deg.C for 2h. The reaction mixture was filtered, the solution extracted with MTBE (10 mL), and the organic layer was discarded. The aqueous phase was then adjusted to ph=2 with 1N HCl, the solution was extracted with EA (50 mL x 5), the combined organic layers were washed with brine (100 mL), and Na 2 SO 4 Drying, filtration, and concentration gave 3-oxo-4, 5-dihydro-1, 4-benzothiazepine-8-carboxylic acid (120 mg,0.487mmol,45% yield) as a white solid.
LCMS(ESI)m/z:[M+H] + =224.1。
1 H NMR(400MHz,DMSO-d 6 )δ=13.09–13.06(m,1H),8.18(t,J=6.4Hz,1H),7.64(d,J=1.6Hz,1H),7.60-7.57(m,1H),7.29(d,J=8.0Hz,1H),4.45(d,J=6.4Hz,2H),3.91(s,2H)ppm。
Step 6 preparation of 1, 3-trioxo-4, 5-dihydro-1λ6, 4-benzothiazepine-8-carboxylic acid
To 3-oxo-4, 5-dihydro-1, 4-benzothiazepine-8-carboxylic acid (50 mg, 223.97. Mu. Mol) in MeOH (0.5 mL) and H 2 To a solution of O (0.5 mL) was added Oxone (275.37 mg, 447.93. Mu. Mol) and the mixture was stirred at 30deg.C for 2h. The reaction mixture was poured into MeOH (5 mL), the solution was filtered, and the filtrate was concentrated to give 1, 3-trioxo-4, 5-dihydro-1λ6, 4-benzothiazepine-8-carboxylic acid (57 mg,223.31 μmol,99.71% yield) as a white solid.
Intermediate 11.4- (2-methoxyethyl) -3-methylsulfonyl-benzoic acid
Step 1 preparation of 2- (4-chloro-2-methylsulfanyl-phenyl) acetic acid
A mixture of 2- (2-bromo-4-chlorophenyl) acetic acid (1 g,4.01 mmol), cuI (763.36 mg,4.01 mmol) and DABCO (899.20 mg,8.02mmol,881.57 uL) in DMSO (10 mL) was stirred at 145℃under N 2 Stirring for 12h in the atmosphere. The reaction mixture was diluted with 1N HCl (300 mL) and filtered. The filtrate was extracted with DCM (300 mLx 2). By anhydrous Na 2 SO 4 The organic layer was dried and the mixture was dried,filtration and concentration gave a residue. The residue was purified by column chromatography (petroleum ether/ethyl acetate=1/0-0/1). The eluate was concentrated to give 2- (4-chloro-2-methylsulfanyl-phenyl) acetic acid (1.5 g, crude) as a yellow solid which was used directly in the next step.
Step 2 preparation of 2- (4-chloro-2-methylsulfonyl-phenyl) acetic acid
To 2- (4-chloro-2-methylsulfanyl-phenyl) acetic acid (500 mg,2.31 mmol) in MeOH (3 mL) and H at 0deg.C 2 To a solution in O (3 mL) was added H 2 Oxone (4.26 g,6.92 mmol) in O (3 mL). The mixture was stirred at 25℃for 12h. With saturated Na 2 SO 3 The reaction mixture was diluted (100 mL), stirred for 10min, and then extracted with DCM (100 mL. Times.3). By anhydrous Na 2 SO 4 The organic layer was dried, filtered, and concentrated to give a residue. The residue was purified by reverse phase (0.1% FA). The eluate was concentrated to give 2- (4-chloro-2-methylsulfonyl-phenyl) acetic acid (200 mg,0.804mol,35% yield) as a white solid. LCMS (ESI) M/z: [ M+H ]] + =248.9。
1 H NMR(400MHz,DMSO-d6)δ=12.62-12.54(m,1H),7.91(d,J=2.0Hz,1H),7.78-7.76(m,1H),7.55(d,J=8.0Hz,1H),4.05(s,2H),3.26(s,3H)ppm。
Step 3 preparation of 2- (4-chloro-2-methylsulfonyl-phenyl) ethanol.
To a solution of 2- (4-chloro-2-methylsulfonyl-phenyl) acetic acid (200 mg, 804.24. Mu. Mol) in THF (4 mL) at 0deg.C was added BH 3 -Me2 S Mixture (10M, 402.12 uL). The mixture was stirred at 25℃for 2h. The reaction mixture was diluted with 1N HCl (10 mL) and extracted with DCM (10 mL). With no Na 2 SO 4 The organic layer was dried with water, filtered, and concentrated to give a residue. The residue was purified by reverse phase (0.1% FA). The eluate was concentrated to give 2- (4-chloro-2-methylsulfonyl-phenyl) ethanol (180 mg, 766.94. Mu. Mol,96% yield) as a colorless oil. LCMS (ESI) M/z: [ M+H ]] + =235.0。
1 H NMR(400MHz,CDCl 3 )δ=8.06(d,J=2.4Hz,1H),7.58-7.55(m,1H),7.42(d,J=8.0Hz,1H),3.97-3.94(m,2H),3.28-3.25(m,2H),3.15(s,3H)ppm。
Step 4 preparation of 4-chloro-1- (2-methoxyethyl) -2-methylsulfonyl-benzene
To a solution of 2- (4-chloro-2-methylsulfonyl-phenyl) ethanol (80 mg, 0.3411 mmol) in DCM (1 mL) was added Ag 2 O (236.97 mg,1.02 mmol) and MeI (241.91 mg,1.70mmol,106 uL). The mixture was stirred at 30℃for 12h. By H 2 The reaction mixture was diluted with O (10 mL) and extracted with DCM (10 mL. Times.2). By anhydrous Na 2 SO 4 The combined organic layers were dried, filtered and concentrated to give a residue which was purified by reverse phase (0.1% FA). The eluate was concentrated to give 4-chloro-1- (2-methoxyethyl) -2-methylsulfonyl-benzene (60 mg,0.241mmol,71% yield) as a yellow solid.
LCMS(ESI)m/z:[M+H] + =248.9。
1 H NMR(400MHz,CDCl 3 )δ=8.06(d,J=2.4Hz,1H),7.55-7.52(m,1H),7.42(d,J=8.4Hz,1H),3.70-3.67(m,2H),3.33-3.29(m,5H),3.15(s,3H)ppm。
Step 5 preparation of 4- (2-methoxyethyl) -3-methylsulfonyl-benzoic acid ((intermediate 11)
4-chloro-1- (2-methoxyethyl) -2-methylsulfonyl-benzene (60 mg,0.241 mmol), K 2 CO 3 (50.0 mg,0.362 mmol), dicyclohexyl (3-dicyclohexylphosphonium propyl) phosphonium; ditetrafluoroborate (14.77 mg, 24.12. Mu. Mol) and Pd (OAc) 2 (2.71 mg, 12.06. Mu. Mol) in DMSO (1 mL) and H 2 The mixture in O (0.2 mL) was degassed and purged 3 times with CO. The mixture was stirred at 100deg.C under CO atmosphere (15 psi) for 3h. The reaction mixture was diluted with MeOH (10 mL), filtered, and the filtrate concentrated to give a residue. The residue was purified by reverse phase (0.1% FA). The eluate was concentrated to remove ACN and lyophilized to give 4- (2-methoxyethyl) -3-methylsulfonyl-benzoic acid (50 mg,0.194mmol,80% yield) as a white solid. LCMS (ESI) M/z: [ M+H ]] + =259.0。
1 H NMR(400MHz,CDCl 3 )δ=8.78(d,J=1.6Hz,1H),8.28-8.26(m,1H),7.61(d,J=8.4Hz,1H),3.77-3.74(m,2H),3.45-3.42(m,2H),3.33(s,3H),3.19(s,3H)ppm。
Intermediate 12.4- (2-methoxyethyl) -3-methylsulfonyl-benzoic acid
Step 1 preparation of 3- [ allyl (t-butoxycarbonyl) sulfamoyl ] -4-vinyl-benzoic acid methyl ester
To a solution of methyl 3- (allylsulfamoyl) -4-vinyl-benzoate (1.2 g,4.27 mmol) (prepared according to the method in FG-A4366) and DMAP (52.11 mg, 426.55. Mu. Mol) in DCM (20 mL) was added TEA (863.24 mg,8.53mmol,1.19 mL) and Boc at 0deg.C 2 O (1.86 g,8.53mmol,1.96 mL). The mixture was stirred at 20℃for 2h. It was poured into water (60 mL) and extracted with DCM (40 mLx 3). The combined organic layers were washed with brine (40 mL), and dried over Na 2 SO 4 Drying, filtering and concentrating. Purification of the residue by flash chromatography on silica gel40g/>Silica gel flash column, eluent of 0-50% ethyl acetate/petroleum ether gradient @50 mL/min). Concentrating the fraction in vacuo to give 3- [ allyl (tert-butoxycarbonyl) sulfamoyl]-4-vinyl-benzoic acid methyl ester (1.5 g,3.93mmol,92% yield) as a yellow oil.
1 H NMR(400MHz,DMSO-d 6 )δ=8.50(d,J=2.0Hz,1H),8.34-8.15(m,1H),7.92(d,J=8.4Hz,1H),7.23-7.00(m,1H),6.01-5.86(m,2H),5.75-5.61(m,1H),5.39-5.14(m,2H),4.38(d,J=4.8Hz,2H),3.91(s,3H),1.13(s,9H)ppm。
Step 2 preparation of benzo [ f ] [1,2] thiazepine-2, 8 (3H) -dicarboxylic acid 2- (tert-butyl) ester 8-methyl ester 1, 1-dioxide
3- [ allyl (t-butoxycarbonyl) sulfamoyl]-4-vinyl-benzoic acid methyl ester (1.5 g,3.93 mmol) and benzylidene- [1, 3-bis (2, 4, 6-trimethylphenyl) imidazolidin-2-ylidene]-dichloro-ruthenium; a mixture of tricyclohexylphosphine (333.85 mg, 393.24. Mu. Mol) in DCM (80 mL) was degassed with N 2 Purging 3 times. Will beThe mixture was N at 25 DEG C 2 Stirring for 2h in the atmosphere. Concentrate to remove DCM. Purification of the residue by flash chromatography on silica gel40g/>Silica gel flash column, eluent of 0-50% ethyl acetate/petroleum ether gradient @50 mL/min). Concentrating the fractions in vacuo to give benzo [ f][1,2]Thiazepine-2, 8 (3H) -dicarboxylic acid 2- (tert-butyl) ester 8-methyl ester 1, 1-dioxide (1.1 g,2.77mmol,70% yield) as a yellow solid. LCMS (ESI) m/z: [ Br ] 79 M+H] + =298.0
1 H NMR(400MHz,DMSO-d 6 )δ=8.43(d,J=1.6Hz,1H),8.32-8.17(m,1H),7.82(d,J=8.0Hz,1H),6.75(d,J=12.8Hz,1H),6.39-6.18(m,1H),4.95-4.57(m,2H),3.92(s,3H),1.11(s,9H)ppm。
Step 3 preparation of 4, 5-Dihydrobenzo [ f ] [1,2] thiazepine-2, 8 (3H) -dicarboxylic acid 2- (tert-butyl) ester 8-methyl ester 1, 1-dioxide
Benzo [ f][1,2]A mixture of thiepine-2, 8 (3H) -dicarboxylic acid 2- (tert-butyl) ester 8-methyl ester 1, 1-dioxide (500 mg,1.41 mmol), pd/C (50 mg,10% purity) in MeOH (10 mL) was degassed with H 2 Purging 3 times. The mixture was subjected to H at 20 ℃ 2 Stirring for 16h in the atmosphere. Filtering, concentrating to obtain 4, 5-dihydrobenzo [ f ]][1,2]2- (tert-butyl) 2-methyl thief-2, 8-dioxidate 1, 1-dioxide (4.1 g,12.27mmol,96% yield) was a yellow oil.
LCMS(ESI)m/z:[Br 79 M+H] + =300.0。
1 H NMR(400MHz,DMSO-d 6 )δ=8.37(d,J=2.0Hz,1H),8.23-8.11(m,1H),7.66(d,J=8.0Hz,1H),4.17-4.06(m,2H),3.90(s,3H),3.32-3.14(m,2H),1.90-1.53(m,2H),1.22(s,9H)ppm。
Step 4 preparation of 2-tert-Butoxycarbonyl-1, 1-dioxo-4, 5-dihydro-3H-1λ6, 2-benzothiazepine-8-carboxylic acid
To 4, 5-dihydrobenzo [ f][1,2]2- (tert-butyl) ester 8-methyl ester 1, 1-dioxide of thiepine-2, 8 (3H) -dicarboxylic acid (250 mg,0.703 mmol) in THF (2.5 mL) and H 2 LiOH.H was added to a solution in O (2.5 mL) 2 O (118.06 mg,2.81 mmol). The mixture was stirred at 25℃for 2h. The pH was adjusted to pH=5 with aqueous HCl (1M) and extracted with EA (40 mLx 3). The combined organic layers were washed with brine (30 mL), and dried over Na 2 SO 4 Drying, filtration and concentration gave 2-tert-butoxycarbonyl-1, 1-dioxo-4, 5-dihydro-3H-1λ6, 2-benzothiazepine-8-carboxylic acid (190 mg,0.473mmol,67% yield) as a white solid. LCMS (ESI) M/z: [ M+H ] ] + =285.9。
Step 5 preparation of 1, 1-dioxo-2, 3,4, 5-tetrahydro-1λ6, 2-benzothiazepine-8-carboxylic acid (intermediate 12)
A mixture of 2-tert-butoxycarbonyl-1, 1-dioxo-4, 5-dihydro-3H-1λ6, 2-benzothiazepine-8-carboxylic acid (180 mg,0.527 mmol) in HCl/dioxane (4M, 3 mL) was stirred at 25℃for 2H. Concentration to remove dioxane gave 1, 1-dioxo-2, 3,4, 5-tetrahydro-1λ6, 2-benzothiazepine-8-carboxylic acid (130 mg, 0.4638 mmol,89% yield, HCl) as a yellow solid.
1 H NMR(400MHz,DMSO-d 6 )δ=8.31(d,J=1.6Hz,1H),8.05-8.00(m,1H),7.57-7.52(m,2H),3.66(br s,2H),3.22(br d,J=3.2Hz,2H),1.91-1.77(m,1H),1.70(br s,2H)ppm。
Intermediate 13.2-methyl-1, 1-dioxo-4, 5-dihydro-3H-1λ6, 2-benzothiazepine-8-carboxylic acid
Step 1 preparation of 4-bromo-3-chlorosulfonylbenzoic acid
4-Bromobenzoic acid (10 g,49.75 mmol) in HSO 3 A mixture of Cl (86.95 g,0.746mol,49.7 mL) was stirred at 100deg.C for 16h. The reaction system was stirred at 120℃for a further 16h. It was poured into ice water (400 mL). A precipitate formed and the mixture was filtered. The filter cake was dried in vacuo to give 4-bromo-3-chlorosulfonylbenzoic acid (11 g,36.72mmol,73% yield) as a grey solid.
LCMS(ESI)m/z:[Br 79 M+H] + =300.0。
1 H NMR(400MHz,DMSO-d 6 )δ=13.96(br s,1H),8.46(d,J=1.6Hz,1H),7.79-7.60(m,2H)ppm。
Step 2 preparation of 4-bromo-3-chlorosulfonylbenzoic acid methyl ester
4-bromo-3-chlorosulfonylbenzoic acid (11 g,36.72 mmol) in SOCl 2 A mixture of (43.69 g,367.25mmol,26.64 mL) was stirred at 80℃for 2h. The mixture was then concentrated to remove SOCl 2 . MeOH (11 mL) was added. The mixture was stirred at 20deg.C for 0.5hr. It was poured into water (600 mL) and extracted with EA (300 mL. Times.3). The combined organic layers were washed with brine (200 mL), and dried over Na 2 SO 4 Drying, filtration and concentration gave methyl 4-bromo-3-chlorosulfonylbenzoate (10 g, crude) as a yellow solid.
LCMS(ESI)m/z:[Br 79 M+H] + =314.8。
1 H NMR(400MHz,DMSO-d 6 )δ=9.31(br s,2H),8.71-8.31(m,1H),7.89-7.62(m,2H),3.86(s,3H)。
Step 3 preparation of 3- (allylsulfamoyl) -4-bromo-benzoic acid methyl ester
To a solution of methyl 4-bromo-3-chlorosulfonylbenzoate (4 g,12.76 mmol) and prop-2-en-1-amine (1.31 g,14.03mmol,1.73mL, HCl) in DCM (40 mL) was added DIEA (6.60 g,51.03mmol,8.89 mL) at 0deg.C. The mixture was then stirred at 25℃for 2h. It was poured into water (100 mL) and extracted with DCM (60 mLx 3). The combined organic layers were washed with brine (60 mL), and dried over Na 2 SO 4 Drying, filtering and concentrating. Purification of the residue by flash chromatography on silica gel80gSilica gel flash column, eluent of 0-50% ethyl acetate/petroleum ether gradient @100 mL/min). The fractions were concentrated in vacuo to give 3- (allylsulfamoyl) -4-bromo-benzoic acid methyl ester (4.1 g,12.27mmol,96% yield) as a white solid。
1 H NMR(400MHz,DMSO-d 6 )δ=8.51-8.42(m,1H),8.29(br s,1H),8.01(d,J=0.8Hz,2H),5.77-5.52(m,1H),5.17-5.06(m,1H),5.03-4.93(m,1H),3.89(s,3H),3.57(br d,J=4.8Hz,2H)ppm。
Step 4 preparation of 3- (allylsulfamoyl) -4-vinyl-benzoic acid methyl ester
Methyl 3- (allylsulfamoyl) -4-bromo-benzoate (3.1 g,9.28 mmol), potassium trifluoroborate (6.21 g,46.38 mmol), di-tert-butyl (cyclopentyl) phosphine; palladium dichloride; iron (604.6 mg,0.928 mmol) and K 3 PO 4 (5.91 g,27.8 mmol) in dioxane (30 mL) and H 2 The mixture in O (6 mL) was degassed with N 2 Purging 3 times. The mixture was heated to 60℃under N 2 Stirring for 16h in the atmosphere. It was poured into water (100 mL) and extracted with EA (60 mL x 3). The combined organic layers were washed with brine (60 mL), and dried over Na 2 SO 4 Drying, filtering and concentrating. Purification of the residue by flash chromatography on silica gel40gSilica gel flash column, eluent of 0-50% ethyl acetate/petroleum ether gradient @80 mL/min). Fractions were concentrated in vacuo to give methyl 3- (allylsulfamoyl) -4-vinyl-benzoate (1.5 g,5.33mmol,57% yield) as a white solid. LCMS (ESI) M/z: [ M+H ]] + =282.1。
1 H NMR(400MHz,CDCl 3 )δ=8.62(d,J=1.6Hz,1H),8.29-8.11(m,1H),7.69(d,J=8.0Hz,1H),7.62-7.47(m,1H),5.92-5.77(m,1H),5.73-5.53(m,2H),5.22-4.97(m,2H),4.84-4.57(m,1H),3.69-3.41(m,2H)ppm。
Step 5 preparation of 3- [ allyl (methyl) sulfamoyl ] -4-vinyl-benzoic acid methyl ester
To 3- (allylsulfamoyl) -4-vinyl-benzoic acid methyl ester (200 mg,0.711 mmol) and K 2 CO 3 To a solution of (196.5 mg,1.42 mmol) in DMF (2 mL) was added MeI (201.81 mg,1.42mmol, 88.5. Mu.L). Will beThe mixture was stirred at 20℃for 3h. It was poured into water (60 mL) and extracted with EA (30 mL. Times.3). The combined organic layers were washed with brine (20 mL) and then with Na 2 SO 4 Drying, filtering and concentrating. Purification of the residue by flash chromatography on silica gel12gSilica gel flash column, eluent of 0-50% ethyl acetate/petroleum ether gradient @50 mL/min). Concentrating the fraction in vacuo to give 3- [ allyl (meth) sulfamoyl ]-4-vinyl-benzoic acid methyl ester (190 mg,0.643mmol,90% yield) as a yellow oil. LCMS (ESI) M/z: [ M+H ]] + =296.0
1 H NMR(400MHz,CDCl 3 )δ=8.56(d,J=1.6Hz,1H),8.26-8.09(m,1H),7.73(d,J=8.4Hz,1H),7.67-7.53(m,1H),5.88-5.77(m,1H),5.76-5.64(m,1H),5.59-5.50(m,1H),5.27-5.16(m,2H),3.96(s,3H),3.75(d,J=6.4Hz,2H),2.75(s,3H)ppm。
Step 6 preparation of methyl 2-methyl-1, 1-dioxo-3H-1λ6, 2-benzothiazepine-8-carboxylate
To 3- [ allyl (methyl) sulfamoyl ]]-4-vinyl-benzoic acid methyl ester (190 mg,0.643 mmol) and benzylidene- [1,3-bis (2, 4, 6-trimethylphenyl) imidazolidin-2-ylidene]-dichloro-ruthenium; tricyclohexylphosphine (benzone- [1,3-bis (2, 4, 6-trimethylphenyl) imidazolidin-2-ylethylene)]-dichoro-ruthenium; a mixture of tricyclohexylphosphine (54.61 mg, 64.33. Mu. Mol) in DCM 10 mL) was degassed with N 2 Purge 3x. The mixture was stirred at 25℃under N 2 Stirring for 2h in the atmosphere. Purification of the residue by flash chromatography on silica gel12gSilica gel flash column, -50% ethyl acetate/petroleum ether gradient eluent @30 mL/min). Concentrating the fractions in vacuo to give 2-methyl-1, 1-dioxo-methyl 3H-1λ6, 2-benzothiazepine-8-carboxylate (130 mg, 0.481 mmol,76% yield) as a white solid. LCMS (ESI) M/z: [ M+H ]] + =268.0。
1 H NMR(400MHz,DMSO-d 6 )δ=8.38(d,J=2.0Hz,1H),8.26-8.10(m,1H),7.78(d,J=8.4Hz,1H),6.72(br d,J=13.2z,1H),6.31-5.96(m,1H),4.45-4.17(m,2H),3.90(s,3H),2.55(s,3H)ppm。
Step 7 preparation of methyl 2-methyl-1, 1-dioxo-4, 5-dihydro-3H-1λ6, 2-benzothiazepine-8-carboxylate
A mixture of 2-methyl-1, 1-dioxo-3H-1λ6, 2-benzothiazepine-8-carboxylic acid methyl ester (130 mg, 0.4816 mmol), pd/C (13 mg,10% purity) in MeOH (4 mL) was degassed with H 2 Purging 3 times. The mixture is then subjected to H at 20 DEG C 2 Stirring for 2h in the atmosphere. Filtration and concentration gave methyl 2-methyl-1, 1-dioxo-4, 5-dihydro-3H-1λ6, 2-benzothiazepine-8-carboxylate (110 mg,0.408mmol,84% yield) as a white solid. LCMS (ESI) M/z: [ M+H ]] + =270.0。
1 H NMR(400MHz,CDCl 3 )δ=8.56(d,J=2.0Hz,1H),8.17-8.03(m,1H),7.38(d,J=7.6Hz,1H),3.95(s,3H),3.92-3.59(m,2H),3.45-3.23(m,2H),2.65(s,3H),1.91-1.80(m,3H)ppm。
Step 8 preparation of 2-methyl-1, 1-dioxo-4, 5-dihydro-3H-1λ6, 2-benzothiazepine-8-carboxylic acid (intermediate 13)
To methyl 2-methyl-1, 1-dioxo-4, 5-dihydro-3H-1λ6, 2-benzothiazepine-8-carboxylate (110 mg,0.408 mmol) in THF (1 mL) and H 2 LiOH.H was added to the solution in O (1 mL) 2 O (68.56 mg,1.63 mmol). The mixture was stirred at 25℃for 2h. Adjust to h=5 with aqueous HCl (1M) and extract with EA (20 ml x 3). The combined organic layers were washed with brine (20 mL) and then with Na 2 SO 4 Drying, filtration, and concentration gave 2-methyl-1, 1-dioxo-4, 5-dihydro-3H-1λ6, 2-benzothiazepine-8-carboxylic acid (80 mg,0.313mmol,77% yield) as a white solid.
LCMS(ESI)m/z:[M+H] + =519.2
1 H NMR(400MHz,DMSO-d 6 )δ=8.27(d,J=1.6Hz,1H),8.14-8.01(m,1H),7.59(d,J=8.0Hz,1H),3.75-3.55(m,2H),3.23(br s,3H),2.55(s,3H),1.83-1.71(m,2H)ppm。
Intermediate 14.4- (difluoromethyl) -3- (methylsulfonyl) benzoic acid
Step 1 preparation of methyl 3-bromo-4- (difluoromethyl) benzoate
To a solution of methyl 3-bromo-4-formylbenzoate (300 mg,1.23 mmol) in DCM (3 mL) was added DAST (596.87 mg,3.70mmol,489.24 uL). The mixture was stirred at 25℃for 1hr. With saturated NaHCO 3 The reaction mixture was diluted with (20 mL) and extracted with DCM (20 mL). By anhydrous Na 2 SO 4 The organic layer was dried, filtered, and concentrated to give a residue. The residue was purified by column chromatography (petroleum ether/ethyl acetate=1/0-3/1). The eluate was concentrated to give methyl 3-bromo-4- (difluoromethyl) benzoate (190 mg, 716.84. Mu. Mol,58% yield) as a yellow oil.
1 H NMR(400MHz,CDCl 3 )δ=8.28(s,1H),8.09(d,J=8.0Hz,1H),7.75(d,J=8.0Hz,1H),7.06-6.79(m,1H),3.96(s,3H)ppm。
Step 2 preparation of 3-bromo-4- (difluoromethyl) benzoic acid
To methyl 3-bromo-4- (difluoromethyl) benzoate (90 mg, 339.56. Mu. Mol) in THF/MeOH/H 2 To a solution of O=2/1/1 (1 mL) was added NaOH (27.16 mg, 679.11. Mu. Mol). The mixture was stirred at 30℃for 2h. The reaction mixture was diluted with 1N HCl (10 mL) and extracted with DCM (10 mL. Times.2). By anhydrous Na 2 SO 4 The combined organic layers were dried, filtered, and concentrated to give 3-bromo-4- (difluoromethyl) benzoic acid (70 mg,278.86 μmol,82% yield) as a yellow oil, which was used directly in the next step.
Step 3 preparation of 4- (difluoromethyl) -3-methylsulfanyl-benzoic acid
3-bromo-4- (difluoromethyl) benzoic acid (50 mg, 0.199mmol), DABCO (44.68 mg, 0.390 mmol,44 uL) and CuI (37.93 mg, 0.199mmol) in DMSO0.5 mL) was stirred at 145℃for 12h. To this mixture was added 1N HCl to adjust ph=5. The mixture was filtered. The filtrate was concentrated to give a residue. The residue was purified by reverse phase (0.1% fa). The eluate was concentrated to give 4- (difluoromethyl) -3-methylsulfanyl-benzoic acid (30 mg,0.137mmol,69% yield) as a yellow solid. LCMS (ESI) M/z: [ M+H ] ] + =218.9。
1 H NMR(400MHz,CDCl 3 )δ=8.10(s,1H),8.02(d,J=8.4Hz,1H),7.75(d,J=8.0Hz,1H),7.15-6.87(m,1H),2.58(s,3H)ppm。
Step 4 preparation of 4- (difluoromethyl) -3-methylsulfonyl-benzoic acid (intermediate 14)
To a solution of 4- (difluoromethyl) -3-methylsulfanyl-benzoic acid (30 mg, 137.48. Mu. Mol) in MeOH (0.5 mL) was added Oxone (169.03 mg, 274.95. Mu. Mol) in H at 0deg.C 2 Mixtures in O (0.5 mL). The mixture was stirred at 25℃for 12h. By H 2 The reaction mixture was diluted with O (10 mL) and extracted with DCM (10 mL). By anhydrous Na 2 SO 4 The organic layer was dried, filtered, and concentrated to give a residue. The residue was purified by reverse phase (0.1% FA). The eluate was concentrated to give 4- (difluoromethyl) -3- (methylsulfonyl) benzoic acid (20 mg, 79.9. Mu. Mol,58% yield) as a white solid.
LCMS(ESI)m/z:[M+H] + =250.9。
Intermediate 15.6-methyl-5- (methylsulfonyl) nicotinic acid
Step 1 preparation of 6-methyl-5- (methylthio) nicotinic acid
To a solution of 5-fluoro-6-methyl-pyridine-3-carboxylic acid methyl ester (300 mg,1.77 mmol) in DMF (2 mL) was added sodium thiomethoxide (320.30 mg,1.95 mmol). The mixture was stirred at 100℃for 16h. The reaction mixture was quenched with HCl (1M) (40 mL) and extracted with EA/meoh=15/1 (40 mL x 5). By anhydrous Na 2 SO 4 The combined organic layers were dried, filtered and concentrated under reduced pressure to give a residue. Reverse phase purification by HPLC (0.1% FA conditions)Residue. The solution was concentrated under reduced pressure to remove MeCN, then lyophilized to give 6-methyl-5- (methylthio) nicotinic acid (200 mg,1.09mmol,62% yield) as a yellow solid.
LCMS(ESI)m/z:[M+H] + =183.9。
1 H NMR(400MHz,DMSO-d 6 )δ=14.14-12.40(m,1H),8.69(d,J=1.6Hz,1H),7.94(d,J=2.0Hz,1H),2.55(s,3H),2.50(s,3H)ppm。
Step 2 preparation of 6-methyl-5- (methylsulfonyl) nicotinic acid (intermediate 15)
To a solution of 6-methyl-5- (methylthio) nicotinic acid (30 mg, 163.73. Mu. Mol) in MeOH (1 mL) was added(150.98 mg, 0.248 mmol) and H 2 O (1 mL). The mixture was stirred at 25℃for 16h. The reaction mixture was dissolved with DMSO (5 mL) and then filtered to give a filtrate. The filtrate was purified by reverse phase HPLC (0.1% FA conditions). The solution was concentrated under reduced pressure to remove MeCN, then lyophilized to give 6-methyl-5- (methylsulfonyl) nicotinic acid (15 mg,67.8 μmol,41% yield) as a white solid. LCMS (ESI) M/z: [ M+H ]] + =216.1.
1 H NMR(400MHz,DMSO-d 6 )δ=15.43-11.63(m,1H),9.17(d,J=2.0Hz,1H),8.61(d,J=2.0Hz,1H),3.36(s,3H),2.90(s,3H)ppm。
Intermediate 16.3-chloro-4-methyl-5-methylsulfonyl-benzoic acid
Step 1 preparation of 3-chloro-5-chlorosulfonyl 4-methylbenzoic acid
A mixture of 3-chloro-4-methylbenzoic acid (1 g,5.86 mmol) in chlorosulfonic acid (10.25 g,87.93mmol,5.85 mL) was stirred at 120℃for 12h. The reaction mixture was added to H at 0deg.C 2 O (20 mL). A white solid precipitated from the mixture. The solid was collected by filtration and dried under reduced pressure to give 3-chloro-5-chlorosulfonyl 4-methylbenzoic acid (1.2 g,4.46mmol,76% yield)Rate) as a white solid.
1 H NMR(400MHz,DMSO-d6)δ=8.30(d,J=2.0Hz,1H),7.85(d,J=2.0Hz,1H),2.63(s,3H)ppm。
Step 2 preparation of 3-chloro-4-methyl-5-methylsulfonyl-benzoic acid (intermediate 16)
Na at 80 DEG C 2 SO 3 (140.51 mg,1.11 mmol) and NaHCO 3 (280.97 mg,3.34mmol,130.08 uL) in H 2 To a solution of O (1.2 mL) was added 3-chloro-5-chlorosulfonyl-4-methylbenzoic acid (300 mg,1.11 mmol). The mixture was stirred at 80℃for 1hr. 2-Bromoacetic acid (309.8 mg,2.23mmol, 161. Mu.L) and NaOH (89.19 mg,2.23 mmol) were then added and the mixture was stirred at 110℃for 12h. By H 2 The reaction mixture was diluted with O (10 mL) and then 1N HCl was added to adjust ph=3. A white solid precipitated from the mixture. The solid was collected by filtration and dried under reduced pressure to give 3-chloro-4-methyl-5-methylsulfonyl-benzoic acid (120 mg,0.483mmol,43% yield) as a white solid. LCMS (ESI) M/z: [ M+H ]] + =248.9
1 H NMR(400MHz,DMSO-d6)δ=8.41(d,J=1.6Hz,1H),8.21(d,J=1.6Hz,1H),3.32(s,3H),2.74(s,3H)ppm。
Intermediate 17.4-chloro-3-fluoro-5-methylsulfonyl-benzoic acid
Step 1 preparation of 4-chloro-3-fluoro-5-methylsulfanyl-benzoic acid
A mixture of methyl 3-bromo-4-chloro-5-fluoro-benzoate (200 mg, 747.72. Mu. Mol), cuI (142.40 mg, 747.72. Mu. Mol) and DABCO (167.8 mg,1.50mmol, 164. Mu.L) in DMSO (2 mL) was N at 145 ℃ 2 Stirring for 12h in the atmosphere. The reaction mixture was filtered. The filtrate was purified by reverse phase HPLC (0.1% fa conditions). The desired fractions were lyophilized to give 4-chloro-3-fluoro-5-methylsulfanyl-benzoic acid (90 mg,0.371mmol,50% yield) as a white solid. LCMS (ESI) M/z: [ M+H ] ] + =220.9. 1 H NMR(400MHz,DMSO-d 6 )δ=7.66-7.56(m,2H),2.59(s,3H)ppm。
Step 2 preparation of 4-chloro-3-fluoro-5-methylsulfonyl-benzoic acid (intermediate 17)
To 4-chloro-3-fluoro-5-methylsulfanyl-benzoic acid (90 mg,0.408 mmol) in H 2 To a solution of O (1 mL) and MeOH (2 mL) was added(501.5 mg,0.816 mmol). The reaction system is carried out at 20 ℃ under N 2 Stirring for 12h in the atmosphere. To the mixture was added saturated Na 2 SO 3 Aqueous solution (5 mL). The mixture was extracted with EA (5 ml x 3). The combined organic layers were washed with brine (10 mL), and dried over Na 2 SO 4 Drying, filtration and concentration gave 4-chloro-3-fluoro-5-methylsulfonyl-benzoic acid (40 mg,0.158mmol,39% yield) as a white solid. LCMS (ESI) M/z: [ M+H ]] + =252.9.
1 H NMR(400MHz,DMSO-d 6 )δ=8.35(s,1H),8.21-8.19(m,1H),3.45(s,3H)ppm。
Intermediate 18.((2-chloro-1, 6-naphthyridin-7-yl) methyl) carbamic acid tert-butyl ester
Step 1.2 preparation of 2-bromo-5-iodo-pyridin-4-amine
NIS (93.6 g,416 mmol) was added to a solution of 2-bromopyridin-4-amine (60 g, 277 mmol) in MeCN (1.5L) at 80 ℃. The reaction mixture was stirred at 80℃for 36h. The reaction mixture was concentrated under reduced pressure to give a residue. With saturated Na 2 SO 3 The residue was diluted (1.5L) and extracted with EA (1.5L. Times.2). The combined organic layers were washed with brine (1L), and dried over Na 2 SO 4 Drying, filtering, and concentrating under reduced pressure to obtain a residue. Purification of the residue by column chromatography (SiO 2 PE/EA=20:3), concentrated under reduced pressure to give 2-bromo-5-iodo-pyridin-4-amine (65 g,217 mmol) as a pale yellow solid.
1 H NMR(400MHz,CDCl 3 )δ=8.31(s,1H),6.79(s,1H),4.75(br s,2H)ppm。
Step 2.preparation of ethyl 3- (4-amino-6-bromo-3-pyridinyl) prop-2-enoate
Propylene-2-enoic acid ethyl ester (45.1 mL, 418 mmol), et 3 N(43.3mL,311mmol)、Pd(OAc) 2 (2.3 g,10.4 mmol) and tris-o-tolylphosphine (6.3 g,20.7 mmol) were added to a solution of 2-bromo-5-iodo-pyridin-4-amine (62 g,207 mmol) in DMF (620 mL). The mixture was stirred at 100℃for 3h. The reaction mixture was diluted with water (4L) and extracted with EA (2L x 2). The combined organic layers were washed with brine (2L) and dried over Na 2 SO 4 Drying, filtering, and concentrating under reduced pressure to obtain a residue. Purification of the residue by column chromatography (SiO 2 PE/EA=20:3), concentrated under reduced pressure to give ethyl 3- (4-amino-6-bromo-3-pyridinyl) prop-2-enoate (50 g,170 mmol) as a pale yellow solid.
LCMS(ESI)m/z:[79BrM+H]+=271.1.
1 H NMR(400MHz,DMSO-d6)δ=8.23(s,1H),7.73(d,J=16.0Hz,1H),6.90-6.67(m,3H),6.52(d,J=16.0Hz,1H),4.18(d,J=7.2Hz,2H),1.25(d,J=7.2Hz,3H)ppm。
Step 3.7 preparation of bromo-1, 6-naphthyridin-2 (1H) -one
Sodium thiomethoxide (24.2 mL,380 mmol) was added to a solution of ethyl 3- (4-amino-6-bromo-3-pyridinyl) prop-2-enoate (40 g,148 mmol) in EtOH (200 mL). The reaction mixture was stirred at 60℃for 2h. The reaction mixture was diluted with water (400 mL) and then neutralized with 1N HCl to ph=7.0. The solids were filtered and the filter cake was washed with water (50 mL). The filter cake was concentrated under reduced pressure to give 7-bromo-1, 6-naphthyridin-2 (1H) -one (22 g,96.8 mmol) as an off-white solid.
LCMS(ESI)m/z:[79BrM+H]+=224.9.
1 H NMR(400MHz,DMSO-d6)δ=12.08(br s,1H),8.65(s,1H),7.99(d,J=9.6Hz,1H),7.36(s,1H),6.62(d,J=9.6Hz,1H)ppm。
Step 4.2 preparation of 2-oxo-1, 2-dihydro-1, 6-naphthyridine-7-carbonitrile
Zinc powder (406.80 mg,6.22 mmol) was added to 7-bromo-1, 6-naphthyridin-2 (1H) -one (7 g,31.1 mmol), zn (CN) 2 (3.95 mL,62.2 mmol) and Pd (dppf) Cl 2 . CH 2 Cl 2 (5.08 g,6.22 mmol) in DMA (140 mL). The reaction mixture was degassed with N 2 Purge 3 times and then stir the mixture at 120 ℃ for 2h. The reaction mixture was diluted with water (200 mL) and extracted with DCM/isopropanol (v/v=3:1) (150 mL x 2). The combined organic layers were filtered, washed with brine (200 mL), and dried over Na 2 SO 4 Drying, filtering, and concentrating under reduced pressure to obtain a residue. Purification of the residue by column chromatography (SiO 2 PE/EA=1:1), concentrated under reduced pressure to give 2-oxo-1, 2-dihydro-1, 6-naphthyridine-7-carbonitrile (3 g,17.5 mmol) as an off-white solid.
1 H NMR(400MHz,DMSO-d6)δ=12.37(br s,1H),8.97(s,1H),8.09(d,J=9.6Hz,1H),7.67(s,1H),6.77(d,J=9.6Hz,1H)ppm。
Step 5 preparation of 2-chloro-1, 6-naphthyridine-7-carbonitrile
2-oxo-1, 2-dihydro-1, 6-naphthyridine-7-carbonitrile (3.0 g,17.5 mmol) and POCl 3 The mixture of (30 mL,323 mmol) was stirred at 80deg.C for 2h. Pouring the reaction mixture into H 2 O (2L) with NaHCO 3 Adjust to ph=7. The solution was extracted with EA (1.5L x 2), the combined organic layers were washed with brine (2L), and dried over Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave 2-chloro-1, 6-naphthyridine-7-carbonitrile (1.1 g,5.76 mmol) as a brown solid. LCMS (ESI) M/z: [ M+H ] ]+=190.1.
1 H NMR(400MHz,DMSO-d6)δ=9.58(d,J=0.8Hz,1H),8.79(d,J=0.8Hz,1H),8.69(s,1H),8.00(d,J=8.8Hz,1H)ppm。
Preparation of (2-chloro-1, 6-naphthyridin-7-yl) methylamine
At-70 ℃ at N 2 DIBAL-H (1M, 329.64mL,2.5 eq) was added dropwise to a solution of 2-chloro-1, 6-naphthyridine-7-carbonitrile (25 g,131.86 mmol) in DCM (1000 mL) under an atmosphere. The reaction mixture was stirred at-70℃for 2h. The reaction mixture was quenched with water (500 mL) and saturated potassium sodium tartrate (1500 mL) and stirred for an additional 30min. The mixture was extracted with DCM: meoh=10:1 (6000 ml x 3). With Na 2 SO 4 The combined organic layers were dried, filtered and concentrated under reduced pressure to give (2-chloro-1, 6-naphthyridin-7-yl) methylamine (51 g, crude) as a brown solid which was used directly in the next step. LCMS (ESI) m/z: [ 35 ClM+H] + =194.2.
Step 7 preparation of tert-butyl ((2-chloro-1, 6-naphthyridin-7-yl) methyl) carbamate (intermediate 18)
To a solution of (2-chloro-1, 6-naphthyridin-7-yl) methylamine (51 g,263.4 mmol) in DCM (1500 mL) was added (Boc) 2 O (172.45 g,790.16 mmol) and DIEA (102.12 g,790.16 mmol). The mixture was stirred at 25℃for 16h. The reaction mixture was diluted with water (1500 mL) and then filtered. The filtrate was extracted with DCM (1000 mL. Times.3). The combined organic layers were washed with brine (1500 mL), and dried over Na 2 SO 4 Drying, filtering, and concentrating under reduced pressure to obtain a residue. Purification of the residue by column chromatography (SiO 2 Petroleum ether/ethyl acetate=10/1-2/1-1/3), concentrating under reduced pressure, eluting to obtain tert-butyl ((2-chloro-1, 6-naphthyridin-7-yl) methyl) carbamate (21 g,64.34mmol,24% yield) as pale yellow solid.
LCMS(ESI)m/z:[M+H]+=293.9.
1 H NMR(400MHz,DMSO-d 6 )δ=9.37(s,1H),8.62(d,J=8.4Hz,1H),7.71(d,J=8.4Hz,1H),7.58-7.53(m,2H),4.39(d,J=6.4Hz,2H),4.20–4.25(m,2H),1.41(s,9H)ppm。
Intermediate 19 [2- [6- (2, 2-difluorocyclopropyl) -2-pyridinyl ] -1, 6-naphthyridin-7-yl ] methylamine
Step 1 preparation of 2-bromo-6- (2, 2-difluorocyclopropyl) pyridine
At 70 ℃ at N 2 TMSCF was added to a mixture of 2-bromo-6-vinylpyridine (500 mg,2.72 mmol) and NaI (81.45 mg,0.543 mmol) in THF (4 mL) in an atmosphere 3 (1.55 g,10.87 mmol) in THF (1 mL) over 1h. The mixture was stirred at 70℃under N 2 Stirring for 1h in the atmosphere. The residue was purified by silica gel chromatography (PE-PE/ea=50/1). The eluate was concentrated under reduced pressure to give 2-bromo-6- (2, 2-difluorocyclopropyl) pyridine (570 mg,2.44mmol,90% yield) as a yellow oil. LCMS (ESI) M/z: [ M+H ]] + =233.9.
1 H NMR(400MHz,CDCl 3 )δ=7.52-7.48(m,1H),7.39-7.37(m,1H),7.19(d,J=7.6Hz,1H),2.95-2.84(m,1H),2.21-2.12(m,1H),1.89-1.83(m,1H)ppm。
Step 2 preparation of [6- (2, 2-difluorocyclopropyl) -2-pyridinyl ] -trimethylstannane
2-bromo-6- (2, 2-difluorocyclopropyl) pyridine (100 mg, 427.28. Mu. Mol), hexamethylditin (279.97 mg, 854.55. Mu. Mol, 177.20. Mu.L) and Pd (PPh) 3 ) 4 (49.37 mg, 42.73. Mu. Mol) in dioxane (2 mL) at 100deg.C under N 2 Stirring for 2h in the atmosphere. The mixture was filtered and concentrated under reduced pressure to give [6- (2, 2-difluorocyclopropyl) -2-pyridinyl ]]Trimethylstannane (170 mg, crude) as a brown oil.
LCMS(ESI)m/z:[M+H] + =320.1。
Preparation of step 3:N- [ [2- [6- (2, 2-difluorocyclopropyl) -2-pyridinyl ] -1, 6-naphthyridin-7-yl ] methyl ] carbamic acid tert-butyl ester
Tert-butyl ((2-chloro-1, 6-naphthyridin-7-yl) methyl) carbamate (50 mg, 170.21. Mu. Mol), [6- (2, 2-difluorocyclopropyl) -2-pyridinyl]Trimethylstannane (163 mg,0.511 mmol) and Pd (PPh) 3 ) 2 Cl 2 (11.95 mg, 17.02. Mu. Mol) in dioxane (1 mL) at 100deg.C under N 2 Stirring for 16h in the atmosphere. The mixture was poured into saturated KF (10 mL) and stirred at 20 ℃ for 30min. The mixture was extracted with EA (10 mL x 3). By anhydrous Na 2 SO 4 The combined organic layers were dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (PE/ea=10/1-EA). Concentrating the eluate under reduced pressure to obtain N- [ [2- [6- (2, 2-difluorocyclopropyl) -2-pyridinyl ]]-1, 6-naphthyridin-7-yl]Methyl group]Tert-butyl carbamate (30 mg, 72.74. Mu. Mol,43% yield) was a yellow solid. LCMS (ESI) M/z: [ M+H ]] + =413.3。
Step 4 preparation of [2- [6- (2, 2-difluorocyclopropyl) -2-pyridinyl ] -1, 6-naphthyridin-7-yl ] methylamine (intermediate 19)
At 0 ℃ to N- [ [2- [6- (2, 2-difluorocyclopropyl) -2-pyridinyl ]]-1, 6-naphthyridin-7-yl]Methyl group]To a mixture of tert-butyl carbamate (30 mg, 72.74. Mu. Mol) in DCM (1 mL) was added TFA (460 mg,4.05mmol,0.3 mL). The mixture was stirred at 30 ℃Stirring for 1hr. Concentrating the mixture under reduced pressure to give [2- [6- (2, 2-difluorocyclopropyl) -2-pyridinyl ] ]-1, 6-naphthyridin-7-yl]Methylamine (31 mg, 72.71. Mu. Mol,100% yield, TFA salt) as a yellow solid. LCMS (ESI) M/z: [ M+H ]] + =313.2。
Intermediate 20- (2, 2-difluorocyclopropyl) pyrimidin-4-yl) -1, 6-naphthyridin-7-yl) methylaminate
Step 1 preparation of 2, 2-difluorocyclopropanecarboxamide
NH at 0deg.C 4 Cl (6.88 g,128.59 mmol) in toluene (50 mL) was added Al (CH) 3 ) 3 (2M, 64.29 mL). The mixture was then stirred at 25℃for 1hr. To this solution was added methyl 2, 2-difluorocyclopropanecarboxylate (3.5 g,25.72 mmol) at 0deg.C, and the solution was stirred at 80deg.C for 12h. A large amount of white precipitate formed. The reaction mixture was cooled to 0 ℃. MeOH (50 mL) was added and then stirred for 10min. The mixture was filtered. The filtrate was concentrated in vacuo to give 2, 2-difluorocyclopropanecarboxamide (3 g, crude) as a white solid, which was used directly.
Step 2 preparation of 2- (2, 2-difluorocyclopropyl) pyrimidin-4-ol
At 25 ℃ at N 2 K was added in one portion to a mixture of 2, 2-difluorocyclopropanecarboxamide (3.00 g,24.97 mmol) in EtOH (40 mL) in an atmosphere 2 CO 3 (6.90 g,49.94 mmol). The mixture was stirred at 25℃for 10min, then ethyl (E) -3-ethoxyacrylate (1.2 g,8.32mmol,1.20 mL) was added at 25 ℃. The mixture was stirred at 75℃for 6h. The reaction mixture was filtered and the filtrate concentrated in vacuo. The mixture was purified by silica gel chromatography (DCM/meoh=20/1). The eluate was concentrated to give 2- (2, 2-difluorocyclopropyl) pyrimidin-4-ol (500 mg,2.90mmol,35% yield) as a white solid.
LCMS(ESI)m/z:[M+H] + =173.2.
1 H NMR(400MHz,CDCl 3 )δ=8.04-7.95(m,1H),6.43-6.35(m,1H),2.84-2.69(m,1H),2.51-2.39(m,1H),2.00-1.88(m,1H)ppm。
Step 3 preparation of 4-chloro-2- (2, 2-difluorocyclopropyl) pyrimidine
At 0 ℃ at N 2 Oxalyl chloride (516 mg,4.07mmol, 356. Mu.L) was added in one portion to a mixture of 2- (2, 2-difluorocyclopropyl) pyrimidin-4-ol (350 mg,2.03 mmol) and DMF (14.9 mg,0.203mmol,15.6 uL) in DCM (6 mL) under an atmosphere. The mixture was stirred at 25℃for 20min. The mixture was added to saturated NaHCO 3 (50 mL). The aqueous phase was extracted with DCM (50 mL. Times.2). The combined organic phases were washed with brine (50 ml x 1), dried over anhydrous Na 2 SO 4 Drying, filtering and concentrating in vacuum. The residue was purified by silica gel chromatography (PE/ea=10/1). The eluate was concentrated to give 4-chloro-2- (2, 2-difluorocyclopropyl) pyrimidine (150 mg,0.787mmol,39% yield) as a pale yellow oil.
LCMS(ESI)m/z:[M+H] + =190.9,192.9。
Step 4 preparation of 2- (2, 2-difluorocyclopropyl) -4- (tributylstannyl) pyrimidine
At 25 ℃ at N 2 Pd (PPh) was added in one portion to a mixture of 4-chloro-2- (2, 2-difluorocyclopropyl) pyrimidine (100 mg,0.525 mmol) and trimethyl (trimethylstannyl) stannane (343.8 mg,1.05mmol, 218. Mu.L) in dioxane (2 mL) in an atmosphere 3 ) 4 (60.63 mg, 52.47. Mu. Mol). The mixture was stirred at 100℃for 2h. The mixture was poured into water (10 mL) and extracted with ethyl acetate (10 mL x 2). The combined organic phases were washed with brine (10 ml x 1), dried over anhydrous Na 2 SO 4 Drying, filtration and concentration in vacuo afforded 2- (2, 2-difluorocyclopropyl) -4- (tributylstannyl) pyrimidine (150 mg, crude) as a yellow oil.
LCMS(ESI)m/z:[M+H] + =320.9。
Step 5 preparation of tert-butyl ((2- (2, 2-difluorocyclopropyl) pyrimidin-4-yl) -1, 6-naphthyridin-7-yl) methyl) carbamate
At 25 ℃ at N 2 To a mixture of 2- (2, 2-difluorocyclopropyl) -4- (tributylstannyl) pyrimidine (147 mg,0.460 mmol) and tert-butyl ((2-chloro-1, 6-naphthyridin-7-yl) methyl) carbamate (90 mg,0.306 mmol) in dioxane (2 mL) in an atmospherePd (PPh) was added at one time 3 ) 2 Cl 2 (21.51 mg, 30.64. Mu. Mol). The mixture was stirred at 100℃for 12h. The mixture was poured into water (30 mL) and extracted with ethyl acetate (20 mL x 2). The combined organic phases were washed with brine (20 mL x 1), dried over anhydrous Na 2 SO 4 Drying, filtering and concentrating in vacuum. The residue was purified by flash chromatography on silica gel (PE/ea=3/1). The eluate was concentrated to give tert-butyl ((2- (2, 2-difluorocyclopropyl) pyrimidin-4-yl) -1, 6-naphthyridin-7-yl) methyl) carbamate (90 mg,0.218mmol,71% yield) as a yellow solid. LCMS (ESI) M/z: [ M+H ]] + =414.0.
1 H NMR(400MHz,DMSO-d6)δ=9.50-9.43(m,1H),9.08-9.01(m,1H),8.84-8.78(m,1H),8.73-8.67(m,1H),8.50-8.44(m,1H),7.86-7.80(m,1H),7.72-7.60(m,1H),4.51-4.42(m,2H),2.30-2.13(m,1H),1.52-1.41(m,9H),1.41-1.21(m,2H)ppm。
Step 6 preparation of (2- (2, 2-difluorocyclopropyl) pyrimidin-4-yl) -1, 6-naphthyridin-7-yl) formate
At 25 ℃ at N 2 To a mixture of tert-butyl ((2- (2, 2-difluorocyclopropyl) pyrimidin-4-yl) -1, 6-naphthyridin-7-yl) methyl) carbamate (90 mg,0.218 mmol) in DCM (1 mL) was added TFA (770.0 mg,6.75mmol, 500. Mu.L) in one portion. The mixture was stirred at 25℃for 30min. The mixture was poured into ice-water (20 mL) and extracted with ethyl acetate (20 mL x 1). The organic phase was discarded. Adding saturated NaHCO to the aqueous phase 3 To adjust ph=8. The aqueous phase was then extracted with ethyl acetate (20 ml x 2). The combined organic phases were washed with brine (10 ml x 1), dried over anhydrous Na 2 SO 4 Drying, filtration and concentration in vacuo afforded (2- (2, 2-difluorocyclopropyl) pyrimidin-4-yl) -1, 6-naphthyridin-7-yl) carboxylate (70 mg, crude) as a pale yellow solid, which was used without purification.
Intermediate 21- [2- [6- (2, 2-difluoro-1-methyl-cyclopropyl) -2-pyridinyl ] -1, 6-naphthyridin-7-yl ] methylamine
/>
Step 1 preparation of 2-bromo-6- (2, 2-difluoro-1-methyl-cyclopropyl) pyridine
At 70 ℃ at N 2 TMSCF3 (287.19 mg,2.02 mmol) was added dropwise to a mixture of 2-bromo-6-isopropenyl-pyridine (100 mg, 504.90. Mu. Mol) and NaI (15.14 mg, 100.98. Mu. Mol) in THF (0.8 mL) in the atmosphere over a period of 30min. The mixture was stirred at 70℃under N 2 Stirring for 30min in the atmosphere. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (PE-PE/ea=20/1). The eluate was concentrated under reduced pressure to give 2-bromo-6- (2, 2-difluoro-1-methyl-cyclopropyl) pyridine (125 mg,0.504mmol,100% yield) as a yellow oil. LCMS (ESI) M/z: [ M+H ]] + =247.9.
1 H NMR(400MHz,CDCl 3 )δ=7.56-7.50(m,1H),7.40-7.37(m,1H),7.30(d,J=7.6Hz,1H),2.28-2.21(m,1H),1.63-1.59(m,3H),1.48-1.41(m,1H)ppm。
Step 2 preparation of [6- (2, 2-difluoro-1-methylcyclopropyl) -2-pyridinyl ] -trimethyl-stannane
2-bromo-6- (2, 2-difluoro-1-methyl-cyclopropyl) pyridine (100 mg, 403.12. Mu. Mol), hexamethyl-ditin (264.15 mg,0.806mmol, 167. Mu.L) and Pd (PPh) 3 ) 4 (46.58 mg, 40.31. Mu. Mol) in dioxane (2 mL) at 100deg.C under N 2 Stirring for 2h in the atmosphere. The mixture was filtered, and the filtrate was concentrated under reduced pressure to give [6- (2, 2-difluoro-1-methylcyclopropyl) -2-pyridinyl ]]Trimethyl-stannane (210 mg, crude) as a dark brown oil. LCMS (ESI) M/z: [ M+H ]] + =334.0。
Preparation of step 3:N- [ [2- [6- (2, 2-difluoro-1-methyl-cyclopropyl) -2-pyridinyl ] -1, 6-naphthyridin-7-yl ] methyl ] carbamic acid tert-butyl ester
Tert-butyl ((2-chloro-1, 6-naphthyridin-7-yl) methyl) carbamate (60 mg,0.204 mmol), [6- (2, 2-difluoro-1-methylcyclopropyl) -2-pyridinyl]Trimethyl-stannane (203.4 mg, 0.313 mmol) and Pd (PPh) 3 ) 2 Cl 2 (14.34 mg, 20.43. Mu. Mol) in dioxane (1 mL) at 100deg.C under N 2 Stirring for 16h in the atmosphere. The mixture was poured into saturated KF (10 mL) and stirred at 20 ℃ for 30min. The mixture was extracted with EA (10 ml x 3). By anhydrous Na 2 SO 4 Drying the combined organic layers, filtering, concentrating under reduced pressure to obtainTo residue. The residue was purified by silica gel chromatography (PE/ea=10/1-EA). Concentrating the eluate under reduced pressure to obtain N- [ [2- [6- (2, 2-difluoro-1-methyl-cyclopropyl) -2-pyridinyl ]]-1, 6-naphthyridin-7-yl]Methyl group]Tert-butyl carbamate (42 mg, 98.49. Mu. Mol,48% yield) was a yellow solid.
LCMS(ESI)m/z:[M+H] + =427.0。
Step 4 preparation of [2- [6- (2, 2-difluoro-1-methyl-cyclopropyl) -2-pyridinyl ] -1, 6-naphthyridin-7-yl ] methylamine (intermediate 21)
To N- [ [2- [6- (2, 2-difluoro-1-methyl-cyclopropyl) -2-pyridinyl ] at 0deg.C]-1, 6-naphthyridin-7-yl]Methyl group]To a solution of tert-butyl carbamate (42 mg, 98.49. Mu. Mol) in DCM (1 mL) was added TFA (462.0 mg,4.05mmol,0.3 mL). The mixture was stirred at 25℃for 1hr. Concentrating the mixture under reduced pressure to give [2- [6- (2, 2-difluoro-1-methyl-cyclopropyl) -2-pyridinyl ]]-1, 6-naphthyridin-7-yl]Methylamine (43 mg, 97.65. Mu. Mol,99% yield, TFA salt) as a yellow solid. LCMS (ESI) M/z: [ M+H ]] + =327.0。
Intermediate 22.1-imino-1-oxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin-8-carboxylic acid
Step 1 preparation of N- (8-bromo-1-oxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin-1-ylidene) -2, 2-trifluoro-acetamide
8-bromo-3, 5-dihydro-2H-4, 1λ4-benzoxathiepin 1-oxide (50 mg, 191.47. Mu. Mol), 2-trifluoroacetamide (64.93 mg, 574.42. Mu. Mol), acetic acid [ acetoxy (phenyl) - λ3-iodo]Esters ([ acetoxy (phenyl) -lambda 3-iodanyl)]A mixture of acetate) (129.51 mg, 402.09. Mu. Mol) and MgO (46.30 mg,1.15 mmol) in DCM (3 mL) was stirred at 25℃for 5min. Then, rhodium diacetoxy (8.46 mg, 19.15. Mu. Mol) was added to the mixture, and the mixture was N-substituted at 25℃in an atmosphere 2 Stirring for 16h. The reaction mixture was diluted with MeOH (3 mL) to give N- (8-bromo-1-oxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin-1-ylidene) -2, 2-trifluoro-acetamide (71 mg, 190.78. Mu. Mol,100% yield) as a yellow liquid for direct useNext, the process is performed. LCMS (ESI) M/z= [ m+h] += 373.2。
Step 2 preparation of 8-bromo-1-imino-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin 1-oxide
To a mixture of N- (8-bromo-1-oxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin-1-ylidene) -2, 2-trifluoro-acetamide (70 mg, 188.09. Mu. Mol) in MeOH (3 mL) was added K 2 CO 3 (181.97 mg,1.32 mmol) and the mixture was stirred at 25℃for 4h. The mixture was diluted with water (10 mL) and filtered to remove the precipitate. The filtrate was separated and the aqueous layer was extracted with DCM (10 mL). The combined organic phases were washed with brine (10 mL), with anhydrous Na 2 SO 4 Drying, filtering, and concentrating the filtrate in vacuo. Purification of the residue by column chromatography (SiO 2 PE: etOAc=20:1-1:1) afforded 8-bromo-1-imino-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin 1-oxide (40 mg,137.65 μmol,73% yield) as a white solid.
LCMS(ESI)m/z=[M+H] + =277.2.
1 H NMR(400MHz,DMSO_d6)δ=8.07(d,J=2.0Hz,1H),7.81-7.79(m,1H),7.46(d,J=8.0Hz,1H),4.99-4.81(m,3H),4.21-4.13(m,2H),3.42-3.39(m,2H)ppm。
Step 3 preparation of 1-imino-1-oxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin-8-carboxylic acid
To 8-bromo-1-imino-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin 1-oxide (40 mg, 144.85. Mu. Mol) and palladium diacetoxy (3.25 mg, 14.48. Mu. Mol) in DMSO (3 mL) and H 2 K was added to the mixture in O (0.3 mL) 2 CO 3 (30.03 mg, 217.27. Mu. Mol) and dicyclohexyl (3-dicyclohexylphosphonium propyl) phosphonium; ditetrafluoroborate (17.74 mg, 28.97. Mu. Mol). The mixture was degassed, purged 3 times with CO, and then stirred at 100 ℃ under a CO atmosphere (15 psi) for 4h. The mixture was poured into water (50 mL), extracted with EA (20.0 mL x 2) and the combined organic layers were discarded. The aqueous layer was adjusted to pH 5 with HCl (1M) and then extracted with DCM (20.0 ml x 3). The combined organic phases were washed with brine (50.0 ml x 2), and dried over Na 2 SO 4 Drying, filtering, evaporating the filtrate to dryness to obtain 1-imino-1-oxo-3, 5-dihydro-2H-4, 1λ6-benzoxepinThiepine-8-carboxylic acid (34 mg, crude) was a yellow solid.
EXAMPLE 2N- [ [2- [6- (azetidin-1-yl) -2-pyridinyl ] -1, 6-naphthyridin-7-yl ] methyl ] -1, 1-dioxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin-8-carboxamide
Preparation of [6- (azetidin-1-yl) -2-pyridinyl ] -trimethyl-stannane
To a solution of 2- (azetidin-1-yl) -6-bromo-pyridine (150 mg, 703.98. Mu. Mol) in dioxane (3 mL) was added hexamethylditin (461.28 mg,1.41 mmol) and Pd (PPh) 3 ) 4 (81.35 mg, 70.40. Mu. Mol). By N 2 The mixture was purged 3x and then at 100 ℃ under N 2 Stirring for 2h in the atmosphere. By H 2 The reaction mixture was diluted with O (200 mL) and extracted with EA (150 mL. Times.3). The combined organic layers were washed with brine (200 mL), and dried over Na 2 SO 4 Drying, filtering, concentrating the filtrate under reduced pressure to give [6- (azetidin-1-yl) -2-pyridinyl ]]Trimethyl-stannane (209 mg, crude) as brown oil was used in the next step without further purification. LCMS (ESI) M/z: [ M+H ]] + =299.3。
Preparation of tert-butyl N- [ [2- [6- (azetidin-1-yl) -2-pyridinyl ] -1, 6-naphthyridin-7-yl ] methyl ] carbamate
To N- [ (2-chloro-1, 6-naphthyridin-7-yl) methyl]To a solution of tert-butyl carbamate (100 mg, 340.43. Mu. Mol) in dioxane (2 mL) was added [6- (azetidin-1-yl) -2-pyridinyl]Trimethyl-stannane (202.2 mg, 680.7. Mu. Mol) and Pd (PPh) 3 ) 2 Cl 2 (23.9 mg, 34.04. Mu. Mol). By N 2 The mixture was purged 3 times and then N at 100deg.C 2 Stirring for 12h in the atmosphere. By H 2 The reaction mixture was diluted with O (20 mL) and extracted with EA (30 mL. Times.3). The combined organic layers were washed with brine (30 mL), and dried over Na 2 SO 4 Drying, filtering, concentrating the filtrate under reduced pressure to obtain a residue, and purifying by column chromatography (SiO 2 Petroleum ether/ethyl acetate=10:1-1:1) to obtain N-/d-(2- (6- (azetidin-1-yl) pyridin-2-yl) -1, 6-naphthyridin-7-yl) methyl) carbamic acid tert-butyl ester (70 mg, 173.45. Mu. Mol,51% yield) as a yellow solid. LCMS (ESI) M/z: [ M+H ] ] + =392.4; 1 H NMR(400MHz,CDCl 3 )δ=9.22(s,1H),8.68(d,J=8.8Hz,1H),8.33(d,J=8.4Hz,1H),7.98(d,J=7.2Hz,1H),7.93(s,1H),7.72-7.61(m,1H),6.43(d,J=8.4Hz,1H),4.68(d,J=4.8Hz,2H),4.18-4.14(m,4H),2.18(s,2H),1.50(s,9H)ppm。
Preparation of [2- [6- (azetidin-1-yl) -2-pyridinyl ] -1, 6-naphthyridin-7-yl ] methylamine
To a solution of tert-butyl N- ((2- (6- (azetidin-1-yl) pyridin-2-yl) -1, 6-naphthyridin-7-yl) methyl) carbamate (70 mg, 178.82. Mu. Mol) in DCM (3 mL) was added TFA (1 mL) at 0deg.C. The mixture was stirred at 25℃for 2h. The reaction mixture was poured into saturated NaHCO 3 Aqueous (30 mL) was extracted with EA (30 mL. Times.3). The combined organic layers were washed with brine (30 mL), and dried over Na 2 SO4 is dried, filtered, and the filtrate is concentrated under reduced pressure to obtain [2- [6- (azetidin-1-yl) -2-pyridyl group]-1, 6-naphthyridin-7-yl]Methylamine (60 mg, crude) was a yellow solid which was used in the next step without further purification.
LCMS(ESI)m/z:[M+H] + =292.4。
Preparation of N- [ [2- [6- (azetidin-1-yl) -2-pyridinyl ] -1, 6-naphthyridin-7-yl ] methyl ] -1, 1-dioxo-3, 5-dihydro-2H-4, 16-benzoxathiepin-8-carboxamide (1)
To a solution of 1, 1-dioxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin-8-carboxylic acid (24.94 mg, 102.97. Mu. Mol) in DCM (1 mL) were added EDCI (21.38 mg, 111.55. Mu. Mol), HOBt (15.07 mg, 111.55. Mu. Mol) and DIEA (33.27 mg, 257.42. Mu. Mol). Then [2- [6- (azetidin-1-yl) -2-pyridinyl ] is added]-1, 6-naphthyridin-7-yl ]Methylamine (25 mg, 85.81. Mu. Mol). The mixture was stirred at 25℃for 2h. By H 2 The reaction mixture was diluted with O (20 mL) and extracted with EA (30 mL x 3). The combined organic layers were washed with brine (30 mL) and with Na 2 SO 4 Drying, filtering, concentrating the filtrate under reduced pressure to give a residue, which was purified by preparative-TLC (SiO 2 DCM: meoh=15:1) to give the crude product. Then by preparing form-HPLC (0.1% FA additive) further purified the crude product. Concentrating the eluate under reduced pressure to remove MeCN and lyophilizing the residue to give N- [ [2- [6- (azetidin-1-yl) -2-pyridinyl ]]-1, 6-naphthyridin-7-yl]Methyl group]-1, 1-dioxo-3, 5-dihydro-2H-4, 16-benzoxathiepin-8-carboxamide (10.21 mg,19.21 μmol,22% yield) as a yellow solid. LCMS (ESI) M/z= [ m+h] + =261.9.
1 H NMR(400MHz,CD 3 OD)δ=9.33(s,1H),8.69-8.63(m,2H),8.62-8.57(m,1H),8.39(s,1H),8.24(d,J=2.0Hz,1H),7.98(s,1H),7.87(d,J=7.2Hz,1H),7.73-7.65(m,2H),6.54(d,J=7.6Hz,1H),5.07(s,2H),4.95(s,2H),4.39-4.34(m,2H),4.17-4.15(m,4H),3.58-3.53(m,2H),2.53-2.40(m,2H)ppm。
The following compounds in table 2 were prepared using standard chemical procedures and methods similar to those used to prepare example 2.
TABLE 2 Compounds of the invention
/>
/>
/>
/>
/>
/>
EXAMPLE 3N- [ [2- [6- (azetidin-1-yl) -2-pyridinyl ] -1, 6-naphthyridin-7-yl ] methyl ] -1, 1-dioxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin-8-carboxamide
Preparation of (6-fluoro-2-pyridinyl) -trimethyl-stannane
To a mixture of 2-bromo-6-fluoro-pyridine (500 mg,2.84 mmol) in dioxane (5 mL) was added trimethyl (trimethylstannyl) stannane (2.79 g,8.52 mmol) and Pd (PPh) 3 ) 4 (328.31 mg, 284.11. Mu. Mol). By N 2 The mixture was purged for 1min and then stirred at 100 ℃ for 2h. Water (20 mL) was added and the mixture extracted with EtOAc (20 mL. Times.2). The combined organic phases were washed with brine (30 mL), dried over anhydrous Na 2 SO 4 Drying, filtration and concentration of the filtrate in vacuo gave (6-fluoro-2-pyridinyl) -trimethyl-stannane (730 mg, crude) as a brown oil. LCMS (ESI) M/z= [ m+h] + =261.9。
Preparation of tert-butyl N- [ [2- (6-fluoro-2-pyridinyl) -1, 6-naphthyridin-7-yl ] methyl ] carbamate
To N- [ (2-chloro-1, 6-naphthyridin-7-yl) methyl]To a mixture of tert-butyl carbamate (300 mg,1.02 mmol) and 6-fluoro-2-pyridinyl) -trimethyl-stannane (530.85 mg,2.04 mmol) in dioxane (6 mL) was added Pd (PPh) 3 ) 2 Cl 2 (71.68 mg, 102.13. Mu. Mol) with N 2 The mixture was purged for 1min. The resulting mixture was stirred at 110℃for 16h. The reaction mixture was then poured into saturated KF (30 mL), stirred for 30min, and the mixture extracted with EtOAc (30 mL x 2). The combined organic phases were washed with brine (40 mL), with anhydrous Na 2 SO 4 Drying, filtering, and concentrating the filtrate in vacuo. The reaction mixture was purified by column chromatography (SiO 2 PE: etOAc=20:1-1:1) to give N- [ [2- (6-fluoro-2-pyridinyl) -1, 6-naphthyridin-7-yl]Methyl group]Tert-butyl carbamate (230 mg, 649.03. Mu. Mol,64% yield) was a yellow solid.
LCMS(ESI)m/z:[M+H] + =355.1.
1 H NMR(400MHz,CDCl 3 )δ=9.26(s,1H),8.63-8.58(m,2H),8.41-8.39(m,1H),8.04-7.98(m,1H),7.94(s,1H),7.09-7.06(m,1H),5.49(br s,1H),4.69(br d,J=5.2Hz,2H),1.50(s,9H)ppm。
Preparation of [2- (6-fluoro-2-pyridinyl) -1, 6-naphthyridin-7-yl ] methylamine
N- [ [2- (6-fluoro-2-pyridinyl) -1, 6-naphthyridin-7-yl ]]Methyl group]A mixture of tert-butyl carbamate (220 mg, 620.81. Mu. Mol) in HCl/dioxane (2 mL) was stirred at 25℃for 1hr. The mixture was evaporated to dryness and the residue was triturated with MTBE (20 ml x 2). The mixture was filtered and the filter cake evaporated to dryness to give [2- (6-fluoro-2-pyridinyl) -1, 6-naphthyridin-7-yl ]]Methylamine (180 mg, crude, HCl) as a yellow solid. LCMS (ESI) M/z: [ M+H ]] + =255.1。
Preparation of N- [ [2- (6-fluoro-2-pyridinyl) -1, 6-naphthyridin-7-yl ] methyl ] -1, 1-dioxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin-8-carboxamide
To [2- (6-fluoro-2-pyridinyl) -1, 6-naphthyridin-7-yl]To a mixture of methylamine (180 mg, 619.15. Mu. Mol) and 1, 1-dioxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin-8-carboxylic acid (180 mg,0.743 mmol) in DCM (2 mL) was added DIEA (320.08 mg,2.48 mmol), EDCI (178 mg, 0.328 mmol) and HOBt (125.49 mg, 928.72. Mu. Mol). The mixture was stirred at 25℃for 1hr. The mixture was poured into water (20 mL) and extracted with EA (10.0 mL. Times.3). The combined organic layers were washed with brine (20.0 mL), and dried over Na 2 SO 4 Drying, filtering and evaporating the filtrate to dryness. The residue was purified by preparative-HPLC (0.1% FA conditions) and the eluate was concentrated in vacuo to remove MeCN. The residue was lyophilized to give N- [ [2- (6-fluoro-2-pyridinyl) -1, 6-naphthyridin-7-yl ] ]Methyl group]-1, 1-dioxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin-8-carboxamide (255 mg,0.528mmol,85% yield)) As a white solid.
LCMS(ESI)m/z:[M+H] + =479.0.
1 H NMR(400MHz,DMSO-d 6 )δ=9.66-9.61(m,1H),9.45(d,J=0.8Hz,1H),8.75-8.72(m,1H),8.54-8.49(m,3H),8.26-8.26(m,1H),8.25-8.17(m,1H),7.84(s,1H),7.73(d,J=8.0Hz,1H),7.39-7.36(m,1H),4.97(s,2H),4.83(d,J=5.6Hz,2H),4.23-4.21(m,2H),3.68-3.66(m,2H)ppm。
Preparation of N- [ [2- [6- [ (2R) -2-methylmorpholin-4-yl ] -2-pyridinyl ] -1, 6-naphthyridin-7-yl ] methyl ] -1, 1-dioxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin-8-carboxamide (37)
To N- [ [2- (6-fluoro-2-pyridinyl) -1, 6-naphthyridin-7-yl]Methyl group]-1, 1-dioxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepine-8-carboxamide (20 mg,0.0418 mmol) and (2R) -2-methylmorpholine; DIEA (27.0 mg,0.209 mmol) was added to a mixture of hydrochloride (17.26 mg, 125.39. Mu. Mol) in DMSO (1 mL). The mixture was stirred at 120℃for 16h. The mixture was then poured into saturated NaHCO 3 (20 mL) was extracted with EA (10.0 mL. Times.3). The combined organic layers were washed with brine (20.0 mL), and dried over Na 2 SO 4 Drying, filtering and evaporating the filtrate to dryness. By preparative HPLC (column: shim-pack C18X 25X 10um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:38% -58%,10 min) the residue was purified and the eluate was concentrated in vacuo to remove MeCN. The residue was lyophilized to give N- [ [2- [6- [ (2R) -2-methylmorpholin-4-yl ]]-2-pyridyl group]-1, 6-naphthyridin-7-yl]Methyl group]-1, 1-dioxo-3, 5-dihydro-2H-4, 1λ6-benzoxathiepin-8-carboxamide (15.89 mg, 26.24. Mu. Mol,63% yield, FA) as a yellow solid.
LCMS(ESI)m/z:[M+H] + =560.3.
1 H NMR(400MHz,DMSO-d 6 )δ=9.65-9.62(m,1H),9.40(s,1H),8.68-8.61(m,2H),8.54(d,J=2.0Hz,1H),8.46(s,1H),8.28-8.25(m,1H),7.92(d,J=7.2Hz,1H),7.81(s,1H),7.77-7.73(m,2H),7.03(d,J=8.4Hz,1H),4.98(s,2H),4.82(d,J=5.6Hz,2H),4.30-4.22(m,4H),3.99-3.96(m,1H),3.70-3.58(m,4H),2.94-2.87(m,1H),2.62-2.56(m,1H),1.22(d,J=6.0Hz,3H)ppm。
The following examples in table 3 were prepared using standard chemical procedures and methods similar to those used to prepare example 3.
TABLE 3 Compounds of the invention
/>
/>
/>
/>
Example 4N- ((2- (4-methyl-3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-8-yl) -1, 6-naphthyridin-7-yl) methyl) -3, 5-dihydro-2H-benzo [ e ] [1,4] oxathiazepin-8-carboxamide 1, 1-dioxide
Step 1 preparation of 4-methyl-8- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 4-dihydro-2H-benzo [ b ] [1,4] oxazine
Pd (dppf) Cl 2 (32.1 mg,0.0448 mmol) and AcOK (129 mg,1.32 mmol) were added to 4, 5-tetramethyl-2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1,3, 2-dioxaborolan (134 mg,0.526 mmol) and 8-bromo-4-methyl-3, 4-dihydro-2H-benzo [ b ]][1,4]Oxazine (100 mg,0.438 mmol) in dioxane (2 mL). The reaction mixture was stirred at 80℃for 2h. By H 2 O (20 mL) dilutes the reactionThe mixture was extracted with EA (20 mL. Times.3). By anhydrous Na 2 SO 4 The combined organic layers were dried, filtered and concentrated under reduced pressure to give the title compound (125 mg, crude) as a brown oil. LCMS (ESI) M/z: [ M+H ]]+=276.1。
Step 2 preparation of tert-butyl ((2- (4-methyl-3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-8-yl) -1, 6-naphthyridin-7-yl) methyl) carbamate
N- [ (2-chloro-1, 6-naphthyridin-7-yl) methyl]Carbamic acid tert-butyl ester (100 mg,0.340 mmol), 4-methyl-8- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 4-dihydro-2H-benzo [ b ]][1,4]Oxazine (122 mg,0.443 mmol), K 3 PO 4 (217 mg,1.02 mmol), [1,1' -bis (di-tert-butylphosphino) ferrocene]Palladium (II) dichloride (22.2 mg,0.340 mmol) in dioxane (1 mL) and H 2 The mixture in O (0.3 mL) was degassed with N 2 Purging 3 times. The mixture was stirred at 80℃for 2h. By H 2 The reaction mixture was diluted with O (10 mL) and extracted with EA (10 mL. Times.3). By anhydrous Na 2 SO 4 The combined organic layers were dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by reverse phase HPLC (0.1% FA additive). The fractions were concentrated under reduced pressure to remove MeCN, then removed with EA (50 ml x 3). By anhydrous Na 2 SO 4 The combined organic layers were dried, filtered and concentrated under reduced pressure to give the title compound (100 mg,0.205 mmol) as a yellow solid. LCMS (ESI) M/z: [ M+H ]]+=407.3. 1 H NMR(400MHz,DMSO-d6)δ=9.30(s,1H),8.48(d,J=8.8Hz,1H),7.98(d,J=8.4Hz,1H),7.69(s,1H),7.63-7.58(m,1H),7.04-7.02(m,1H),6.94-6.90(m,1H),6.87-6.81(m,1H),4.43(d,J=5.6Hz,2H),4.35-4.27(m,2H),3.34-3.33(m,2H),2.91(s,3H),1.43(s,9H)ppm。
Step 3 preparation of (2- (4-methyl-3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-8-yl) -1, 6-naphthyridin-7-yl) methylamine hydrochloride
HCl/dioxane (4 n,750 ul) was added to a solution of tert-butyl ((2- (4-methyl-3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-8-yl) -1, 6-naphthyridin-7-yl) methyl) carbamate (90 mg,0.221 mmol) in dioxane (1 mL). The reaction mixture was stirred at 25℃for 1hr. The reaction mixture was concentrated under reduced pressure. The resulting residue was washed with MTBE (5 ml x 2), filtered and dried under vacuum to give the title compound (70 mg,0.204 mmol) as a brown solid. LCMS (ESI) M/z: [ m+h ] +=307.2.
Step 4 preparation of N- ((2- (4-methyl-3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-8-yl) -1, 6-naphthyridin-7-yl) methyl) -3, 5-dihydro-2H-benzo [ e ] [1,4] oxathiazepin-8-carboxamide 1, 1-dioxide (58)
EDCI (25.2 mg,0.131 mol), HOBt (17.7 mg,0.131 mmol), DIEA (76.2 uL,0.438 mmol) and (2- (4-methyl-3, 4-dihydro-2H-benzo [ b ])][1,4]Oxazin-8-yl) -1, 6-naphthyridin-7-yl) methylamine hydrochloride (30 mg,0.0875 mmol) was added to 2, 3-dihydro-5H-benzo [ e ]][1,4]Oxathiepine-8-carboxylic acid 1, 1-dioxide (25.4 mg,0.105 mmol) in DCM (0.5 mL). The reaction mixture was stirred at 25℃for 2h. By H 2 The reaction mixture was diluted with O (5 mL) and extracted with DCM (5 mL. Times.3). By anhydrous Na 2 SO 4 The combined organic layers were dried, filtered and concentrated under reduced pressure. The resulting residue was purified by reverse phase HPLC (0.1% FA conditions). The solution was concentrated under reduced pressure to remove MeCN and lyophilized to give the title compound (14.2 mg,0.0257 mmol) as a yellow solid. LCMS (ESI) M/z: [ M+H ]]+=531.2. 1 H NMR(400MHz,CD 3 OD)δ=9.30(s,1H),8.64-8.59(m,1H),8.48(d,J=8.8Hz,1H),8.22-8.20(m,1H),8.00(d,J=8.4Hz,1H),7.93(s,1H),7.63(d,J=7.6Hz,1H),7.01-6.92(m,2H),6.88-6.83(m,1H),5.04(s,2H),4.92(s,2H),4.35-4.31(m,4H),3.53-3.50(m,2H),3.34(d,J=4.4Hz,2H),2.97-2.92(m,3H)ppm。
The following examples in table 4A were prepared using standard chemical procedures and methods similar to those used to prepare example 4.
TABLE 4A Compounds of the invention
/>
/>
/>
/>
The following examples in table 4B were prepared using standard chemical procedures and methods similar to those described above.
TABLE 4B Compounds of the invention
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
Example 5 determination of ATPase catalytic Activity of BRM and BRG-1
By using ADP-Glo TM In vitro biochemical assays of (Promega, V9102) to measure the ATPase catalytic activity of BRM or BRG-1. Once the reaction is complete, ADP-Glo is carried out in two steps TM Kinase assay. The first step is to deplete the reaction system of any unconsumed ATP. The second step is to convert the reaction product ADP to ATP, which will be utilized by the luciferase to produce luminescence and detected by a luminescence reader such as Envision.
Assay reaction mixtures (10. Mu.L) containing 30nM BRM or BRG-1, 20nM salmon sperm DNA (from Invitrogen, ultraPure) TM Salmon Sperm DNA Solution, catalog No. 15632011) and 400. Mu.M ATP in ATPase assay buffer containing 20mM Tris, pH 8, 20mM MgCl 2 50mM NaCl, 0.1% Tween-20 and 1mM fresh DTT (Pierce) TM DTT (dithiothreitol), catalog No. 20290). The reaction was initiated by adding 2.5. Mu.L of ATPase solution to 2.5. Mu.L of ATP/DNA solution on a low volume white Proxiplate-384+ plate (Perkinelmer, cat. No. 6008280) and incubating for 1 hour at room temperature. Then, 5. Mu.L of ADP-Glo provided in the addition kit TM After the reagents, the reaction system was incubated at room temperature for 40 minutes. Then, 10 μl of the kinase detection reagent provided in the kit was added to convert ADP to ATP, and the reaction system was incubated at room temperature for 60 minutes. Finally, luminescence measurements are collected using a plate reading photometer, such as Envision.
BRM and BRG-1 are synthesized by high five insect cell lines with purities greater than 90%. IC from atpase catalytic activity assays described herein 50 The data are shown in tables 5A and 5B below.
TABLE 5A BRM and BRG-1 inhibition data for the compounds of the invention
/>
/>
/>
* Ratio of BRG1 IC 50 (mu M) divided by BRM IC 50 (mu M) resulting in a value.
TABLE 5B BRM and BRG-1 inhibition data for the compounds of the invention
/>
/>
/>
/>
/>
/>
EXAMPLE 6 Synthesis of Compound A
BRG1/BRM inhibitor compound a has the structure:
compound a was synthesized as shown in scheme 1 below.
Scheme 1 Synthesis of Compound A
By using the above ADP-Glo TM In vitro biochemical assay of (Promega, V9102) the ATPase catalytic activity of BRM or BRG-1 in the presence of Compound A was determined. Compound a was found to have an IC of 10.4nM for BRM in this assay 50 With 19.3nM for BRG1IC 50
Example 7 effect of BRG1/BRM ATPase inhibition on the growth of uveal melanoma and hematological cancer cell lines
The method comprises the following steps: the uveal melanoma cell line (92-1, MP41, MP38, MP 46), prostate cancer cell Line (LNCAP), lung cancer cell line (NCI-H1299) and immortalized embryonic kidney cell line (HEK 293T) were plated in 96-well plates containing growth medium (see Table 6). BRG1/BRM atpase inhibitor compound a was dissolved in DMSO and added to cells at the time of plating at a concentration gradient of 0 to 10 micromolar. Cells were incubated at 37 degrees celsius for 3 days. After 3 days of treatment, the medium was removed from the cells, and 30 μl of TrypLE (Gibco) was added to the cells for 10 minutes. Cells were separated from the plates and resuspended by adding 170 microliters of growth medium. Cells from both DMSO-treated control wells were counted and the initial cell number plated at the start of the experiment was re-plated in plates containing fresh compound at 37 degrees celsius for another 4 days. On day 7, cells were harvested as described above. On days 3 and 7, relative Cell growth was measured by adding Cell-titer glo (Promega) and luminescence was measured on an Envision plate reader (Perkin Elmer). The concentration of 50% inhibition of growth per cell line was calculated using Graphpad Prism (GI 50 ) And is plotted as follows. For multiple myeloma cell lines (OPM 2, MM1S, LP 1), ALL cell lines (tal 1, JURKAT, RS 411), DLBCL cell lines (sudal 6, sudal 4, DB, WSUDLCL2, PFEIFFER), AML cell lines (ociamal 5), MDS cell lines (SKM 1), ovarian cancer cell lines (OV 7, TYKNU), esophageal cancer cell lines (KYSE 150), rhabdoid tumor lines (RD, G402, G401, HS729, a 204), liver cancer cell lines (HLF, HLE, PLCRPF 5) and lung cancer cell lines (SW 1573, NCIH 2444), the above methods can be performed using the following modifications: cells were plated in 96-well plates, and the next day BRG1/BRM atpase inhibitor compound a was dissolved in DMSO and added to the cells at a concentration gradient of 0 to 10 micromolar. On day 3 and day 7 cells were allowed to divide into new 96-well plates and fresh compound was added 4 hours after re-plating.
Table 6 lists the cell lines tested and the growth media used.
TABLE 6 cell lines and growth media
Cell lines Source Growth medium
92-1 SIGMA RPMI1640+20%FBS
A204 ATCC McCoy's5A+10%FBS
DB ATCC RPMI1640+10%FBS
G401 ATCC McCoy's5A+10%FBS
G402 ATCC McCoy's5A+10%FBS
HEK293T ATCC DMEM+10%FBS
HLE JCRB DMEM+10%FBS
HLF JCRB DMEM+10%FBS
HS729 ATCC DMEM+10%FBS
JURKAT ATCC RPMI1640+10%FBS
KYSE150 DSMZ RPMI1640/Ham'sF12+10%FBS
LNCAP ATCC RPMI1640+10%FBS
LP1 DSMZ IMDM+20%FBS
MM1S ATCC RPMI1640+10%FBS
MP38 ATCC RPMI1640+20%FBS
MP41 ATCC RPMI1640+20%FBS
MP46 ATCC RPMI1640+20%FBS
NCIH1299 ATCC RPMI1640+10%FBS
NCIH2444 ATCC RPMI1640+20%FBS
OCIAML5 DSMZ alpha-MEM+20%FBS+10ng/mlGM-CSF
OPM2 DSMZ RPMI1640+10%FBS
OV7 ECACC DMEM/Ham's F12 (1:1) +2mM glutamine+10% FBS+0.5ug/ml hydrocortisone+10 ug/ml insulin
PFEIFFER ATCC RPMI1640+10%FBS
PLCPRF5 ATCC EMEM+10%FBS
RD ATCC DMEM+10%FBS
RS411 ATCC RPMI1640+10%FBS
SKM1 JCRB RPMI1640+10%FBS
SUDHL4 DSMZ RPMI1640+10%FBS
SUDHL6 ATCC RPMI1640+20%FBS
SW1573 ATCC DMEM+10%FBS
TALL1 JCRB RPMI1640+10%FBS
TYKNU JCRB EMEM+20%FBS
WSUDLCL2 DSMZ RPMI1640+10%FBS
Results: as shown in fig. 1, the uveal melanoma and hematological cancer cell lines were more sensitive to BRG1/BRM inhibition than the other test cell lines. Inhibition of uveal melanoma and hematological cancer cell lines was maintained until day 7.
Example 8 comparison of BRG1/BRM inhibitor with clinical PKC and MEK inhibitors in uveal melanoma cell lines
The method comprises the following steps: the uveal melanoma cell line 92-1 or MP41 was plated in 96-well plates in the presence of growth medium (see Table 5). BAFATP enzyme inhibitors (Compound A), PKC inhibitors (LXS 196; medChemexpress) or MEK inhibitors (semtinib; selleck Chemicals) were dissolved in DMSO and added to cells at the time of plating in a concentration gradient of 0 to 10. Mu. Mol. Cells were incubated at 37 degrees celsius for 3 days. After 3 days of treatment, cell growth was measured by cell titer luminescence (Promega) and luminescence read on an Envision plate reader (Perkin Elmer).
Results: as shown in fig. 2A and 2B, compound a showed comparable growth inhibition of uveal melanoma cells as clinical PKC and MEK inhibitors. Furthermore, compound a was found to produce a more rapid onset of inhibition than clinical PKC and MEK inhibitors.
EXAMPLE 9 Synthesis of Compound B
BRG1/BRM inhibitor compound B has the structure:
compound B was synthesized as shown in scheme 2 below.
Scheme 2: synthesis of Compound B
Preparation of (S) -1- (methylsulfonyl) -N- (4- (methylsulfanyl) -1-oxo-1- ((4- (3- (pyridin-4-yl) phenyl) thiazol-2-yl) amino) butan-2-yl) -1H-pyrrole-3-carboxylate (Compound B)
To (2S) -2-amino-4-methylsulfanyl-N- [4- [3- (4-pyridinyl) phenyl ]]Thiazol-2-yl]To a mixture of butyramide (2 g,4.75mmol, HCl salt) and 1-methylsulfonylpyrrole-3-carboxylic acid (898.81 mg,4.75 mmol) in DMF (20 mL) was added EDCI (1.37 g,7.13 mmol), HOBt (962.92 mg,7.13 mmol) and DIEA (2.46 g,19.00mmol,3.31 mL) and the mixture was stirred at 25℃for 3 h. Pouring the mixture into H 2 O (100 mL) and the precipitate was collected by filtration. The solid was triturated in MeOH (20 mL) and the precipitate collected by filtration. The solid was dissolved in DMSO (10 mL), then the mixture was poured into MeOH (50 mL), the precipitate formed collected by filtration, and lyophilized to give compound B (2.05 g,3.66mmol,77.01% yield) as a white solid. LCMS (ESI) M/z [ M+H] + =555.9. 1 H NMR(400MHz,DMSO)δ12.49(s,1H),8.68-8.66(m,2H),8.46(d,J=7.2Hz,1H),8.31-8.30(m,1H),8.02-8.00(m,1H),7.94-7.96(m,1H),7.83(s,1H),7.73-7.74(m,3H),7.61-7.57(m,1H),7.31-7.29(m,1H),6.79-6.77(m,1H),4.74-4.69(m,1H),3.57(s,3H),2.67-2.53(m,2H),2.13-2.01(m,5H).SFC:AS-3-MeOH(DEA)-40-3mL-35T.lcm,t=0.932min,ee%=100%.
Example 10 effect of BRG1/BRM ATPase inhibition on the growth of uveal melanoma, hematological cancer, prostate cancer, breast cancer and Ewing's sarcoma cell line
The method comprises the following steps: all cell lines described in example 7 above were also tested with compound B as described above. In addition, the following cell lines were also tested as follows. Briefly, for the Ewing sarcoma cell line (CADOES 1, RDES, SKES 1), retinoblastoma cell line (WERIRB 1), ALL cell line (REH), AML cell line (KASUMI 1), prostate cancer cell line (PC 3, DU145,22RV 1), melanoma cell line (SH 4, SKMEL28, WM115, COLO829, SKMEL3, A375), breast cancer cell line (MDAMB 415, CAMA1, MCF7, BT474, HCC1419, DU4475, BT 549), B-ALL cell line (SUPB 15), CML cell line (K562, MEG01, burkitt lymphoma cell line (RAMOS 2G64C10, DAUDI), mantle cell lymphoma cell line (JE 1, REC 1), bladder cancer cell line (HT 1197) and lung cancer cell line (SBC 5), the above method was modified by plating cells in 96-well plates, adding BRG1/BRM inhibitors to fresh cell gradients at a fresh BRM 6 m/BRM 10 and adding to a fresh cell gradient of the well plates at a new cell concentration of 96-well plates at a new BRM 4 m.
Table 7 lists the test cell lines and the growth media used.
TABLE 7 cell lines and growth media
Cell lines Source Growth medium
22RV1 ATCC RPMI1640+10%FBS
A375 ATCC DMEM+10%FBS
BT474 ATCC Hybrid medium+1.5g/L sodium bicarbonate+10% FBS
BT549 ATCC RPMI1640+0.023IU/ml insulin+10% FBS
CADOES1 DSMZ RPMI1640+10%FBS
CAMA1 ATCC EMEM+10%FBS
COLO829 ATCC RPMI1640+10%FBS
DAUDI ATCC RPMI1640+10%FBS
DU145 ATCC EMEM+10%FBS
DU4475 ATCC RPMI1640+10%FBS
HCC1419 ATCC RPMI1640+10%FBS
HT1197 ATCC EMEM+10%FBS
JEKO1 ATCC RPMI1640+20%FBS
K562 ATCC IMDM+10%FBS
KASUMI1 ATCC RPMI1640+10%FBS
MCF7 ATCC EMEM+0.01mg/ml bovine insulin+10% FBS
MDAMB415 ATCC Leibovitz's L-15+2mM L-Glutamine+10 mcg/ml insulin+10 mcg/ml glutathione+15% FBS
MEG01 ATCC RPMI1640+10%FBS
PC3 ATCC F-12K+10%FBS
RAMOS2G64C10 ATCC RPMI1640+10%FBS
RDES ATCC RPMI1640+15%FBS
REC1 ATCC RPMI1640+10%FBS
REH ATCC RPMI1640+10%FBS
SBC5 JCRB EMEM+10%FBS
SH4 ATCC DMEM+10%FBS
SKES1 ATCC McCoy's 5A+15%FBS
SKMEL28 ATCC EMEM+10%FBS
SKMEL3 ATCC McCoy's 5A+15%FBS
SUPB15 ATCC IMDM+4mM L-glutamine+1.5 g/L sodium bicarbonate+0.05 mM 2-mercaptoethanol+20% FBS
WERIRB1 ATCC RPMI1640+10%FBS
WM115 ATCC EMEM+10%FBS
Results: as shown in fig. 3, the eye pigment layer melanoma, hematological cancer, prostate cancer, breast cancer, and ewing's sarcoma cell lines were more sensitive to BRG1/BRM inhibition than the other test cell lines. Inhibition of uveal melanoma, hematological cancer, prostate cancer, breast cancer, and ewing's sarcoma cell lines continued until day 7.
EXAMPLE 11 effect of BRG1/BRM ATPase inhibition on growth of cancer cell lines
The method comprises the following steps: pooled cell viability assays were performed as described previously ("High-throughput identification of genotype-specific cancervulnerabilities in mixtures of barcoded tumor cell lines", yu et al, nature Biotechnology 34,419-423,2016) using the following modifications. Cell lines were obtained from the collection Cancer Cell Line Encyclopedia (CCLE) and were adapted to RPMI-1640 medium without phenol red supplemented with 10% heat-inactivated Fetal Bovine Serum (FBS) in order to adapt the unique infection and pooling protocol to such a huge array of cell lines. A lentiviral rotainfection protocol was performed to introduce a 24 nucleotide barcode into each cell line using blasticidin (blast) as a selectable marker with an estimated fold infection (MOI) of 1 for all cell lines. Then, over 750 stable barcoded PRISM cancer cell lines were pooled together at doubling time, 25 per pool. For screening performance, instead of plating 25 cell line pools in each well as previously described (Yu et al), all adherent cells or all suspension cell lines were plated together using T25 flasks (100000 cells/flask) or 6 well plates (50000 cells/well), respectively. Cells were treated with DMSO or compound starting from the highest concentration of 10 μm in triplicate with 8-point 3-fold dose response. As a control for determining stability, cells were treated in parallel with two previously validated compounds, namely the pan-Raf inhibitor AZ-628 and the proteasome inhibitor bortezomib, at a maximum concentration of 2.5 μm and 0.039 μm, respectively.
After 3 days of treatment with the compounds, the cells were lysed, genomic DNA was extracted, the barcodes were amplified by PCR and detected by Next-Generation Sequencing. Cell viability was determined by comparing the count of cell line specific barcodes in the treated samples to the count of cell line specific barcodes in DMSO control and day 0 control. The dose-response curve for each cell line was fitted and the corresponding area under the curve (AUC) calculated and compared to the median AUC for all cell lines (fig. 4). Cell lines with AUC less than the median are considered to be most sensitive.
Example 12 effect of brg1/BRM atpase inhibitors on ocular pigment layer melanoma cell lines.
The method comprises the following steps: the uveal melanoma cell line (92-1, MP41, MP38, MP 46) and non-small cell lung cancer cells (NCIH 1299) were plated in 96-well plates containing growth medium (see Table 6). BRG1/BRM atpase inhibitor compound 67 was dissolved in DMSO and added to cells at the time of plating at a concentration gradient of 0 to 10 micromolar. Cells were incubated at 37℃for 3 days. After 3 days of treatment, cell growth was measured by cell titer luminescence (Promega) and luminescence read on an Envision plate reader (Perkin Elmer).
Results: as shown in fig. 5, compound B resulted in effective growth inhibition in the eye pigment layer melanoma cell line.
Example 13 comparison of BRG1/BRM inhibitor with clinical PKC and MEK inhibitors in uveal melanoma cells
The method comprises the following steps: the uveal melanoma cell line 92-1 or MP41 was plated in 96-well plates in the presence of growth medium (see Table 6). BAF atpase inhibitor (compound B), PKC inhibitor (LXS 196; medChemExpress) and MEK inhibitor (semetinib; selleck Chemicals) were dissolved in DMSO and added to cells at the time of plating in a concentration gradient of 0 to 10 micromolar. Cells were incubated at 37℃for 3 days. After 3 days of treatment, cell growth was measured by cell titer luminescence (Promega) and luminescence read on an Envision plate reader (Perkin Elmer).
Results: as shown in fig. 6A and 6B, compound B showed more potent effect on growth inhibition of uveal melanoma cells compared to clinical PKC and MEK inhibitors. In addition, compound B was found to initiate growth inhibition faster than clinical PKC and MEK inhibitors.
EXAMPLE 14 BRG1/BRM ATPase inhibitor is effective to inhibit the growth of PKC inhibitor-resistant cells
The method comprises the following steps: MP41 eye pigment layer melanoma cells develop resistance to PKC inhibitors (LXS 196; medChemexpress) by prolonged culture in growth medium containing increasing concentrations of up to 1 micromolar of the compound (see Table 6). After 3 months, parent MP41 cells and PKC inhibitor (PKCi) -resistant cells were tested for sensitivity to either PKC inhibitor (LXS 196) or BRG1/BRM ATPase inhibitor (Compound B) in a 7-day growth inhibition assay as described in example 9 above.
Results: although PKCi resistant cells can tolerate growth at higher concentrations of LXS196 than the parental MP41 cell line (fig. 7A), BRG1/BRM atpase inhibitors (compound B) still resulted in strong growth inhibition of PKCi resistant and parental cell lines (fig. 7B). PKCi resistant cells were more sensitive to compound B than parental MP41 cells (fig. 7B).
EXAMPLE 15 Synthesis of Compound C
Step 1.6 preparation of fluoropyridine-2-carbonyl chloride (intermediate B)
To a cooled (0deg.C) solution of 6-fluoropyridine-2-carboxylic acid (50.00 g,354.36 mmol) in dichloromethane (500 mL) and N, N-dimethylformamide (0.26 mL,3.54 mmol) was added oxalyl chloride (155.10 mL,1.77 mol). After the oxalyl chloride addition was complete, the reaction mixture was warmed to room temperature and stirred for an additional 0.5h. The mixture was concentrated in vacuo to afford intermediate B (56.50 g) as a white solid which was used in the next step without further purification.
Step 2.2 preparation of 2-chloro-1- (6-fluoro-2-pyridinyl) ethanone (intermediate C)
To a cooled (0 ℃) mixture of intermediate B (56.00 g,351.00 mmol) in 1, 4-dioxane (800 mL) was added dropwise a 2M solution of trimethylsilyl diazomethane in hexane (351 mL). The resulting reaction mixture was stirred at 25℃for 10h. The reaction mixture was then quenched with 4M HCl in 1, 4-dioxane (500 mL). After stirring for 2h, the reaction solution was concentrated in vacuo to give an oil. With saturated NaHCO 3 The residue was diluted with aqueous solution (500 mL) and extracted with ethyl acetate (200 mL x 3). The combined organic layers were washed with brine (300 ml x 2), and dried over Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave intermediate C (35.50 g) as a white solid which was used directly in the next step. LCMS (ESI) M/z: [ M+H ]] + =173.8.
Step 3 preparation of 4- (6-fluoro-2-pyridinyl) thiazol-2-amine (intermediate E)
Intermediate C (35.50 g,204.53 mmol) and thiourea (14.01 g,184.07 mmol) in MeOH (250 mL) and H at room temperature 2 NaF (3.56 g,84.82 mmol) was added to a solution of O (250 mL) in a mixture. After stirring for 0.5h, the reaction mixture was partially concentrated under vacuum to remove MeOH and the resulting solution was acidified to pH 3 with 2M aqueous HCl. After 15min, the solution was extracted with ethyl acetate (200 mL. Times.3), the organic layer was discarded, and NaHCO was used 3 (500 mL) alkalizing the aqueous phase, stirring for 30min, then extracting with ethyl acetate (325 mL x 3), washing the combined organic layers (225 mL x 3) with brine, and washing with Na 2 SO 4 Drying, filtering, and concentrating under reduced pressure. The residue was triturated with petroleum ether (300 mL), stirred at 25℃for 10min, and filtered. The resulting solid was dried in vacuo to afford intermediate E (28.00 g,143.43mmol,70.13% yield, 100% purity) as a white solid. LCMS (ESI) M/z: [ M+H ]] + =195.8.; 1 H NMR(400MHz,DMSO-d 6 )δ8.00-7.96(m,1H),7.72(d,J=7.2Hz,1H),7.24(s,1H),7.16(s,2H),7.02(d,J=8.0Hz,1H)。
Preparation of tert-butyl N- [2- [ [4- (6-fluoro-2-pyridinyl) thiazol-2-yl ] amino ] -2-oxo-ethyl ] carbamate (intermediate G)
To a solution of N-Boc-glycine (5.92 g,33.81 mmol), HATU (12.86 g,33.81 mmol) and DIEA (15.89 g,122.94mmol,21.41 mL) in dichloromethane (100 mL) was added intermediate E (6.00 g,30.74 mmol). After stirring for 2h, the reaction mixture was concentrated, then diluted with water (100 mL) and extracted with ethyl acetate (60 mL x 4). The combined organic layers were washed with brine (100 ml x 2), and dried over Na 2 SO 4 Drying, filtering, and concentrating under reduced pressure to obtain a residue. The residue was triturated with a mixture of 1:1 petroleum ether and MeOH (40 mL). After stirring at 25 ℃ for 20min, the suspension was filtered, the filter cake was washed with MTBE (20 mL) and dried in vacuo to afford intermediate G (7.7G, 21.63mmol,70.4% yield, 99.0% purity) as a white solid. LCMS (ESI) M/z: [ M+H ] ] + =353.1.
Step 5 preparation of 2- ((4- (6-fluoropyridin-2-yl) thiazol-2-yl) amino) -2-oxoethyl-1-ammonium chloride (intermediate H)
A solution of intermediate G (5.40G, 15.32 mmol) in 4M HCl in 1, 4-dioxane (35 mL) was stirred at 25deg.C for 1.5h. The mixture was concentrated in vacuo to afford intermediate H (4.42 g) as a white solid which was used directly in the next step without further purification. LCMS (ESI) M/z: [ M+H ]] + =252.9。
Step 6.preparation of 1-tert-butyl-N- [2- [ [4- (6-fluoro-2-pyridinyl) thiazol-2-yl ] amino ] -2-oxo-ethyl ] pyrrole-3-carboxamide (intermediate J)
To a solution of intermediate H (3.00 g,10.39 mmol), 1-tert-butylpyrrole-3-carboxylic acid (1.74 g,10.39 mmol) and DIEA (6.71 g,51.95mmol,9.05 mL) in dichloromethane (40 mL) was added HOBt (1.68 g,12.47 mmol) and EDCI (2.39 g,12.47 mmol) in sequence. After stirring for 4h, the mixture was concentrated in vacuo. The residue was diluted with water (250 mL) and extracted with ethyl acetate (200 mL x 3). The combined organic layers were washed with brine (300 ml x 3), and dried over Na 2 SO 4 Drying, filtering, and concentrating under reduced pressure. The resulting solid was triturated with a 1:1 MTBE/ethyl acetate mixture (400 mL) and after 30min the suspension was filtered. The solid was washed with MTBE (85 ml x 3) and then dried in vacuo to afford intermediate J (3.10 g,7.64mmol,73.6% yield, 99.0% purity) as a white solid. LCMS (ESI) M/z: [ M+H ] ] + =402.3. 1 H NMR(400MHz,DMSO-d 6 )δ12.40(s,1H),8.18-8.15(m,1H),8.09-8.08(m,1H),7.87-7.83(m,2H),7.52(s,1H),7.11(d,J=8.0Hz,1H),6.97(m,1H),6.47(s,1H),4.10(d,J=5.6Hz,2H),1.49(s,9H).
Step 7.preparation of 1- (tert-butyl) -N- (2- ((4- (6- (cis-2, 6-dimethylmorpholino) pyridin-2-yl) thiazol-2-yl) amino) -2-oxoethyl) -1H-pyrrole-3-carboxylate (Compound C)
To a solution of intermediate J (0.100 g, 0.247 mmol) in DMSO (1 mL) was added DIEA (0.130 mL,0.747 mmol) and cis-2, 6-dimethylmorpholine (0.057 g,0.498 mmol), and the mixture was stirred at 120 ℃. After 12h, the solution was cooled to room temperature and the reaction mixture was diluted with MeOH (3 mL). By preparative-HPLC (0.1% TFA; column: luna C18:25.5 u; mobile phase: [ water (0.075% TFA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:30% -60%,2 min) purification residue. The appropriate fractions were collected and lyophilized to give compound C (0.079 g,0.129mmol,51.94% yield, 100% purity) as a white solid. LCMS (ESI) M/z: [ M+H ]] + =497.5;
1 H NMR(400MHz,DMSO-d 6 )δ12.27(s,1H),8.17-8.14(m,1H),7.75(s,1H),7.63-7.59(m,1H),7.51(s,1H),7.25(d,J=7.2Hz,1H),6.96(s,1H),6.79(d,J=8.8Hz,1H),6.47(s,1H),4.24(d,J=12.4Hz,2H),4.08(d,J=5.6Hz,2H),3.64-3.61(m,2H),2.44-2.38(m,2H),1.49(s,9H),1.18(d,J=5.6Hz,6H)。
Example 16 inhibition of in vivo ocular pigment layer melanoma growth by BRG1/BRM ATPase inhibitors
The method comprises the following steps: subcutaneous implantation of 5x10 into armpit area of nude mice (Envigo) 6 92-1 eye pigment layer melanoma cells. Grow the tumor to 200mm 3 Mean, at which time mice were grouped and dosing was started. Mice were given either vehicle (20% 2-hydroxypropyl- β -cyclodextrin) or increasing doses of compound C once daily by oral gavage. Tumor volume and body weight were measured over the course of 3 weeks and the dose was adjusted according to body weight to achieve the appropriate dose in mg/kg. At this point, the animals were sacrificed, tumors were dissected and imaged.
Results: tumor growth inhibition was caused in a dose-dependent manner with compound C, and tumor regression was observed at the highest (50 mg/kg) dose. (FIGS. 8A and 8B). All treatments were well tolerated and no weight loss was observed (fig. 8C).
Other embodiments
While the application has been described in connection with specific embodiments thereof, it will be understood that the application is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the application following, in general, the principles of the application and including such departures from the present disclosure as come within known or customary practice within the art to which the application pertains and as may be applied to the essential features hereinbefore set forth, and as follows in the scope of the appended claims.
Other embodiments are in the claims.

Claims (117)

1. A compound having the structure:
wherein the method comprises the steps of
m is 0, 1, 2 or 3;
n is 0, 1, 2, 3 or 4;
X 1 is-S-, -SO 2 -or-S (O) (NH) -;
X 2 is N or CR 8
R 1 Is hydrogen or optionally substituted C 1 -C 6 An alkyl group;
each R 2 And each R 3 Independently hydrogen, optionally substituted C 1 -C 6 Alkyl or optionally substituted C 1 -C 6 A heteroalkyl group;
L 1 is an optionally substituted 9-or 10-membered bicyclic heterocyclyl or an optionally substituted 9-or 10-membered bicyclic heteroaryl;
L 2 Absent, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted C 6 -C 10 Aryl, optionally substituted 5-to 14-membered heteroaryl, or optionally substituted 4-to 14-membered heterocyclyl;
R 4 is hydrogen, halogen, optionally substituted C 1 -C 6 Alkyl or optionally substituted C 3 -C 10 Cycloalkyl;
R 5 is optionally substituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl or optionally substituted amino, and R 6 Is hydrogen, halogen, cyano, optionally substituted C 1 -C 6 Alkyl, optionally substituted C 2 -C 6 Alkenyl or optionally substituted C 3 -C 10 Cycloalkyl; or R is 5 And R is 6 And together with the attached atoms, are combined into an optionally substituted 5-to 8-membered heterocyclyl;
each R 7 Independently optionally substituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl, halogen, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted C 3 -C 10 Cycloalkyl C 1 -C 6 Alkyl, optionally substituted 5-to 14-membered heteroaryl, optionally substituted 4-to 14-membered heterocyclyl, -N (R) 7A ) 2 OR-OR 7A Wherein each R is 7A Independently H, optionally substituted C 1 -C 6 Alkyl, optionally substituted C 1 -C 6 Heteroalkyl, optionally substituted C 3 -C 10 Cycloalkyl, optionally substituted C 6 -C 10 Aryl, optionally substituted 5-to 10-membered heteroaryl or optionally substituted 4-to 10-membered heterocyclyl, or two geminal R 7A The groups are combined with the attached atoms to form an optionally substituted 5-to 10-membered heteroaryl or an optionally substituted 4-to 10-membered heterocyclyl; or two twin R 7 The groups are combined with the attached atoms to form carbonyl groups;
R 8 is hydrogen, halogen, optionally substituted C 1 -C 6 Alkyl or optionally substituted C 3 -C 10 Cycloalkyl; and is also provided with
R 9 Hydrogen or halogen;
or a pharmaceutically acceptable salt thereof.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 5 And R is 6 And are combined with the attached atoms to form an optionally substituted 5-to 8-membered heterocyclic group.
3. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 5 And R is 6 And are combined with the attached atoms to form an optionally substituted 7-membered heterocyclic group.
4. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 5 Is optionally substituted C 1 -C 6 An alkyl group.
5. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 5 Is an optionally substituted amino group.
6. The compound of claim 1, 4 or 5, or a pharmaceutically acceptable salt thereof, wherein R 6 Is optionally substituted C 1 -C 6 An alkyl group.
7. The compound of claim 1, 4 or 5, or a pharmaceutically acceptable salt thereof, wherein R 6 Is halogen.
8. The compound of any one of claims 1-7, or a pharmaceutically acceptable salt thereof, wherein X 1 Is SO 2
9. The compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof, wherein X 2 Is CR (CR) 8
10. The compound of claim 1, or a pharmaceutically acceptable salt thereof, whereinA group of the structure->Wherein the method comprises the steps of
Z is CH 2 CO or C (R) X2 ) 2
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups;
each R X2 Independently H or optionally substituted C 1 -C 6 An alkyl group; and is also provided with
p is 0, 1, 2, 3 or 4.
11. The compound of claim 1, or a pharmaceutically acceptable salt thereof, whereinA group of the structure->Wherein the method comprises the steps of
Z is CH 2 CO or C (R) X2 ) 2
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups;
each R X2 Independently H or optionally substituted C 1 -C 6 An alkyl group; and is also provided with
p is 0, 1, 2, 3 or 4.
12. The compound of claim 1, or a pharmaceutically acceptable salt thereof, whereinA group of the structure->Wherein the method comprises the steps of
Z is CH 2 CO or C (R) X2 ) 2
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups;
each R X2 Is independently hydrogen or optionally substituted C 1 -C 6 An alkyl group;
p is 0, 1, 2, 3 or 4; and is also provided with
q is 0 or 1.
13. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein A group of the structure->Wherein the method comprises the steps of
Is a single bond or a double bond;
each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups;
R X2 is hydrogen or optionally substituted C 1 -C 6 An alkyl group; and is also provided with
p is 0, 1, 2, 3 or 4.
14. The compound of claim 1, or a pharmaceutically acceptable salt thereof, whereinA group of the structure->Wherein the method comprises the steps of
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups;
R X2 is hydrogen or optionally substituted C 1 -C 6 An alkyl group; and is also provided with
p is 0, 1, 2, 3 or 4.
15. The compound of any one of claims 1-14, or a pharmaceutically acceptable salt thereof, wherein R 8 Is hydrogen.
16. The compound of any one of claims 1-14, or a pharmaceutically acceptable salt thereof, wherein R 8 Is halogen.
17. The compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof, wherein X 2 Is N.
18. The compound of claim 1, or a pharmaceutically acceptable salt thereof, whereinA group of the structure->Wherein the method comprises the steps of
Z is CH 2 CO or C (R) X2 ) 2
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups;
Each R X2 Is independently hydrogen or optionally substituted C 1 -C 6 An alkyl group; and is also provided with
p is 0, 1, 2, 3 or 4.
19. The compound of claim 1, or a pharmaceutically acceptable salt thereof, whereinA group of the structure->Wherein the method comprises the steps of
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The radicals being combined with the atoms to which they are attached to form carbonyl groups or C 3 -C 8 Cycloalkyl ring, or two ortho-positions R X1 The radicals being combined with the atoms to which they are attached to form C 3 -C 8 A cycloalkyl ring;
p is 0, 1, 2, 3 or 4; and is also provided with
q is 0, 1 or 2.
20. The compound of claim 19, or a pharmaceutically acceptable salt thereof, whereinA group of the structure->Wherein the method comprises the steps of
Each R X1 Independently optionally substituted C 1 -C 6 Alkyl or halogen, or two geminal R X1 The groups are combined with the attached atoms to form carbonyl groups; and is also provided with
p is 0, 1, 2, 3 or 4.
21. The compound of any one of claims 10-14 and 18-20, or a pharmaceutically acceptable salt thereof, wherein at least one R X1 Is optionally substituted C 1 -C 6 Alkyl, or at least one R X1 Is halogen.
22. The compound of any one of claims 10-14 and 18-21, or a pharmaceutically acceptable salt thereof, wherein at least two geminal R X1 The groups combine with the attached atoms to form carbonyl groups.
23. The compound of any one of claims 1-22, or a pharmaceutically acceptable salt thereof, wherein L 1 Is an optionally substituted 9-or 10-membered bicyclic heteroaryl.
24. The compound of claim 23, or a pharmaceutically acceptable salt thereof, wherein L 1 Is that
Wherein the method comprises the steps of
X 3 、X 4 、X 5 、X 6 、X 7 And X 8 Each independently is N or CR L1
Each R L1 Independently H, halogen, optionally substituted C 1 -C 6 An alkyl group;
A 1 is bonded to- (C (R) 2 )(R 3 )) m -a bond; and is also provided with
A 2 Is bonded to L 2 Is a key of (c).
25. Weight(s)The compound of claim 24, or a pharmaceutically acceptable salt thereof, wherein L 1 Is that
26. The compound of claim 24, or a pharmaceutically acceptable salt thereof, wherein L 1 Is that
27. The compound of claim 24, or a pharmaceutically acceptable salt thereof, wherein L 1 Is that
28. The compound of claim 24, or a pharmaceutically acceptable salt thereof, wherein L 1 Is that
29. The compound of claim 24, or a pharmaceutically acceptable salt thereof, wherein L 1 Is that
30. The compound of claim 23, or a pharmaceutically acceptable salt thereof, wherein L 1 Is thatWherein A is 1 Is bonded to- (C (R) 2 )(R 3 )) m -a bond; and A is 2 Is bonded to L 2 Is a key of (c).
31. Claims 1 to 30A compound of any one of claims, or a pharmaceutically acceptable salt thereof, wherein L 2 Is an optionally substituted 5-to 14-membered heteroaryl.
32. The compound of any one of claims 1-30, or a pharmaceutically acceptable salt thereof, wherein-L 2 -(R 7 ) n Is a group of the structure:
33. the compound of any one of claims 1-30, or a pharmaceutically acceptable salt thereof, wherein-L 2 -(R 7 ) n Is a group of the structure:
34. the compound of claim 33, or a pharmaceutically acceptable salt thereof, wherein-L 2 -(R 7 ) n Is a group of the structure:
35. the compound of claim 32, or a pharmaceutically acceptable salt thereof, wherein-L 2 -(R 7 ) n Is a group of the structure:
36. the compound of claim 32, or a pharmaceutically acceptable salt thereof, wherein-L 2 -(R 7 ) n Is a group of the structure:
37. the compound of claim 32, or a pharmaceutically acceptable salt thereof, wherein-L 2 -(R 7 ) n Is a group of the structure:
38. the compound of claim 32, or a pharmaceutically acceptable salt thereof, wherein-L 2 -(R 7 ) n Is a group of the structure:
39. the compound of claim 32, or a pharmaceutically acceptable salt thereof, wherein-L 2 -(R 7 ) n Is a group of the structure:
40. the compound of claim 32, or a pharmaceutically acceptable salt thereof, wherein-L 2 -(R 7 ) n Is a group of the structure:
41. the compound of claim 32, or a pharmaceutically acceptable salt thereof, wherein-L 2 -(R 7 ) n Is a knot as followsThe structural group:
42. the compound of any one of claims 1-30, or a pharmaceutically acceptable salt thereof, wherein L 2 Is optionally substituted C 6 -C 10 Aryl groups.
43. The compound of any one of claims 1-42, or a pharmaceutically acceptable salt thereof, wherein n is 1.
44. The compound of any one of claims 1-42, or a pharmaceutically acceptable salt thereof, wherein n is 2.
45. The compound of any one of claims 1-42, or a pharmaceutically acceptable salt thereof, wherein n is 3.
46. The compound of any one of claims 1-45, or a pharmaceutically acceptable salt thereof, wherein R 7 Is optionally substituted C 1 -C 6 An alkyl group.
47. The compound of any one of claims 1-46, or a pharmaceutically acceptable salt thereof, wherein R 7 Is optionally substituted C 1 -C 6 A heteroalkyl group.
48. The compound of any one of claims 1-47, or a pharmaceutically acceptable salt thereof, wherein R 7 Is an optionally substituted 4-to 10-membered heterocyclyl.
49. The compound of claim 48, or a pharmaceutically acceptable salt thereof, wherein R 7 Is optionally substituted azetidinyl or optionally substituted morpholinyl.
50. The compound of any one of claims 1-49 or a pharmaceutically acceptable salt thereofWherein R is 7 Is optionally substituted C 3 -C 10 Cycloalkyl groups.
51. The compound of claim 50, or a pharmaceutically acceptable salt thereof, wherein R 7 Is optionally substituted cyclopropyl or optionally substituted cyclobutyl.
52. The compound of any one of claims 1-51, or a pharmaceutically acceptable salt thereof, wherein R 7 is-N (R) 7A ) 2
53. The compound of claim 52, or a pharmaceutically acceptable salt thereof, wherein R 7 Is optionally substituted N-azetidinyl or optionally substituted N-morpholinyl.
54. The compound of any one of claims 1-53, or a pharmaceutically acceptable salt thereof, wherein two geminal R 7 The groups are combined with the attached atoms to form an optionally substituted 4-to 10-membered heterocyclic group.
55. The compound of any one of claims 1-54, or a pharmaceutically acceptable salt thereof, wherein at least one R 7 is-OR 7A
56. The compound of claim 52, or a pharmaceutically acceptable salt thereof, wherein R 7A Is optionally substituted C 1-6 An alkyl group.
57. The compound of any one of claims 1-42, or a pharmaceutically acceptable salt thereof, wherein n is 0.
58. The compound of any one of claims 1-56, or a pharmaceutically acceptable salt thereof, wherein at least one R 7 Is cyclopropyl, 2-difluorocyclopropyl, difluoromethoxy, 2, 6-dimethylmorpholin-4-yl, N-azetidinyl, 3-fluorocyclobutyl, 2-methoxyethyl, ethoxy, methoxy, 2-difluoroethoxy, 2-difluoroethyl, trifluoromethyl, isopropyl, methylAcetyl, fluoro, chloro, 1-methylpyrazol-3-yl, dimethylamino, N-methyl-N- (2-methoxyethyl) -amino, N-ethyl-N- (2-methoxyethyl) -amino, N- (2-propyl) -N- (2-methoxyethyl) -amino, 2-methoxyethylamino, 3-aza-8-oxa-bicyclo [4.3.0 ] ]Non-3-yl, 3-aza-7-oxa-bicyclo [4.3.0]Non-3-yl, 1-fluorocyclobutan-1-yl, 3-fluoropyrrolidin-1-yl, 3-methoxypyrrolidin-1-yl, oxetan-3-yl, N-methylindolin-4-yl, 2-difluoro-3-methylcyclopropan-1-yl, 3-methoxyazetidin-1-yl, 3-methoxypiperidin-1-yl, 1, 2-dimethyl-7-azaindol-4-yl, 1-methyl-7-azaindol-4-yl, 2, 3-methylenedioxyphenyl, N-methyl-N- (3-oxetan) amino, 3-oxetan-oxy, 1-difluoro-5-azaspiro [2.3 ]]Hex-5-yl, 1-fluoromethyl-cyclopropyl, N- (3-tetrahydrofuranyl) methylamino, N-indolinyl, N-1, 4-oxaazepinyl, 2-fluoro-2-propyl, 1-difluoro-2-propyl, 2-difluoro-1-methylcyclopropan-1-yl, 1-methylcyclopropyl, 4-difluoropiperidin-1-yl, 2-methoxyethoxy, 3-difluorocyclobutan-1-yl, N-methyl-N-1-methoxyprop-2-ylamino 1-methoxyprop-2-ylamino, 1-methoxyethyl, 4-methylpiperazinyl, 3-methylmorpholine, 2-difluoropropoxy, 3-methoxycyclobutyl, methylamino, 4-dimethylamino-3, 3-difluoropiperidinyl, 4-methylamino-3, 3-difluoropiperidinyl, 3-difluoropyrrolidinyl, N-methyl-N-3-methoxycyclobutylamino, 1-methylpyrazol-5-yl, 6-oxa-3-azabicyclo [3.1.1 ]Hept-3-yl, cyclopropoxy, 2, 6-dimethylpyridin-4-yl, 2-methylpyrrolidinyl, 4-oxabicyclo [4.1.0 ]]Hept-1-yl, N-methyl-N- (2, 6-dimethyltetrahydropyran-4-yl) amino or N-methyl-N-3-methyloxetan-3-ylmethylamino.
59. The compound of any one of claims 1-58, or a pharmaceutically acceptable salt thereof, wherein R 1 Is hydrogen.
60. A compound selected from compounds 1-308 and pharmaceutically acceptable salts thereof.
61. A compound selected from compounds 309-856 and pharmaceutically acceptable salts thereof.
62. The compound of any one of claims 1-61, or a pharmaceutically acceptable salt thereof, wherein the compound has a BRG1 IC of at least 5 50 And BRM IC 50 Is a ratio of (2).
63. The compound of any one of claims 1-61, or a pharmaceutically acceptable salt thereof, wherein the compound has a BRG1 IC of at least 7 50 And BRM IC 50 Is a ratio of (2).
64. The compound of any one of claims 1-61, or a pharmaceutically acceptable salt thereof, wherein the compound has a BRG1 IC of at least 10 50 And BRM IC 50 Is a ratio of (2).
65. The compound of any one of claims 1-61, or a pharmaceutically acceptable salt thereof, wherein the compound has a BRG1 IC of at least 15 50 And BRM IC 50 Is a ratio of (2).
66. The compound of any one of claims 1-61, or a pharmaceutically acceptable salt thereof, wherein the compound has a BRG1 IC of at least 20 50 And BRM IC 50 Is a ratio of (2).
67. The compound of any one of claims 1-61, or a pharmaceutically acceptable salt thereof, wherein the compound has a BRG1 IC of at least 25 50 And BRM IC 50 Is a ratio of (2).
68. The compound of any one of claims 1-61, or a pharmaceutically acceptable salt thereof, wherein the compound has a BRG1 IC of at least 30 50 And BRM IC 50 Is a ratio of (2).
69. A pharmaceutical composition comprising a compound of any one of claims 1-68 and a pharmaceutically acceptable excipient.
70. A method of reducing BAF complex activity in a cell, the method comprising contacting the cell with an effective amount of a compound of any one of claims 1-68 or a pharmaceutical composition of claim 69.
71. The method of claim 70, wherein the BAF complex is in a cancer cell.
72. A method of treating a BAF complex-associated disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of claims 1-68 or the pharmaceutical composition of claim 69.
73. The method of claim 72, wherein the BAF complex associated disorder is cancer or a viral infection.
74. A method of inhibiting BRM, the method comprising contacting a cell with an effective amount of a compound of any one of claims 1-68 or a pharmaceutical composition of claim 69.
75. The method of claim 73, wherein the cell is a cancer cell.
76. A method of treating a disorder involving a BRG1 loss-of-function mutation in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of any one of claims 1-68 or a pharmaceutical composition of claim 69.
77. The method of claim 76, wherein the disorder involving a loss of BRG1 function mutation is cancer.
78. A method of inducing apoptosis in a cell comprising contacting the cell with an effective amount of a compound of any one of claims 1-68 or a pharmaceutical composition of claim 69.
79. The method of claim 78, wherein the cell is a cancer cell.
80. A method of treating cancer in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of any one of claims 1-68 or a pharmaceutical composition of claim 69.
81. The method of any one of claims 71-80, wherein the cancer is non-small cell lung cancer, colorectal cancer, bladder cancer, primary unknown cancer, glioma, breast cancer, melanoma, non-melanoma skin cancer, endometrial cancer, esophageal gastric cancer, pancreatic cancer, hepatobiliary cancer, soft tissue sarcoma, ovarian cancer, head and neck cancer, renal cell carcinoma, bone cancer, non-hodgkin's lymphoma, small cell lung cancer, prostate cancer, embryonic carcinoma, germ cell tumor, cervical cancer, thyroid cancer, salivary gland cancer, gastrointestinal neuroendocrine tumor, uterine sarcoma, gastrointestinal stromal tumor, CNS cancer, thymoma, adrenal cortex cancer, appendiceal cancer, small intestine cancer, or penile cancer.
82. The method of claim 81, wherein the cancer is non-small cell lung cancer, colorectal cancer, bladder cancer, primary unknown cause cancer, glioma, breast cancer, melanoma, non-melanoma skin cancer, endometrial cancer, or penile cancer.
83. The method of claim 82, wherein the cancer is non-small cell lung cancer.
84. The method of claim 82, wherein the cancer is soft tissue sarcoma.
85. A method of treating a viral infection in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of any one of claims 1-68 or a pharmaceutical composition of claim 69.
86. The method of claim 85, wherein the viral infection is a viral infection of the retrovirus family, hepadnaviridae family, flaviviridae family, adenovirus family, herpesviridae family, papillomaviridae family, parvoviridae family, picoviridae family, polyomaviridae family, paramyxoviridae family, or togaviridae family.
87. A method of treating melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematological cancer in a subject in need thereof, comprising administering to the subject an effective amount of a compound of any one of claims 1-68 or a pharmaceutical composition of claim 69.
88. A method of reducing tumor growth of melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematological cancer in a subject in need thereof, comprising administering to the subject an effective amount of the compound of any one of claims 1-68 or the pharmaceutical composition of claim 69.
89. A method of inhibiting the progression of metastasis of melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematological cancer in a subject, the method comprising administering an effective amount of the compound of any one of claims 1-68 or the pharmaceutical composition of claim 69.
90. A method of inhibiting metastatic colonization of melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma or hematological cancer in a subject, the method comprising administering an effective amount of a compound of any one of claims 1-68 or a pharmaceutical composition of claim 69.
91. A method of reducing BRG1 and/or BRM levels and/or activity in a melanoma, prostate, breast, bone, renal cell or hematological cancer cell, the method comprising contacting the cell with an effective amount of a compound of any one of claims 1-68 or a pharmaceutical composition of claim 69.
92. The method of claim 91, wherein the cell is in a subject.
93. The method of any one of claims 87-92, wherein the melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematological cancer is metastatic.
94. The method of any one of claims 87-92, wherein the method further comprises administering an anti-cancer therapy to the subject or contacting the cells with an anti-cancer therapy.
95. The method of claim 94, wherein the anti-cancer therapy is a chemotherapeutic or cytotoxic agent, immunotherapy, surgery, radiation therapy, thermal therapy, or photocoagulation, or a combination thereof.
96. The method of claim 95, wherein the anti-cancer therapy is surgery.
97. The method of claim 95, wherein the anti-cancer therapy is a chemotherapeutic agent or a cytotoxic agent.
98. The method of claim 97, wherein the chemotherapeutic or cytotoxic agent is an antimetabolite, an antimitotic, an antitumor antibiotic, an asparagine-specific enzyme, a bisphosphonate, an antitumor agent, an alkylating agent, a DNA-repair enzyme inhibitor, a histone deacetylase inhibitor, a corticosteroid, a demethylating agent, an immunomodulator, a janus-related kinase inhibitor, a phosphatidylinositol 3-kinase inhibitor, a proteasome inhibitor, or a tyrosine kinase inhibitor, or a combination thereof.
99. The method of claim 97 or 98, wherein the one or more chemotherapeutic or cytotoxic agents is dacarbazine, temozolomide, cisplatin, trosoxivas, fotemustine, IMCgp100, CTLA-4 inhibitors, PD-1 inhibitors, PD-L1 inhibitors, mitogen-activated protein kinase inhibitors, and/or protein kinase C inhibitors.
100. The method of any one of claims 94-99, wherein the anti-cancer therapy and the compound of any one of claims 1-38 or the pharmaceutical composition of claim 39 are administered within 28 days of each other and each in an amount effective together to treat the subject.
101. The method of any one of claims 87-100, wherein the subject or cancer has and/or has been identified as having a BRG1 loss-of-function mutation.
102. The method of any one of claims 87-101, wherein melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematologic cancer fails to respond to administration or progresses after administration of one or more chemotherapeutic or cytotoxic agents.
103. The method of any one of claims 87-102, wherein the melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematologic cancer is resistant to or is expected to be resistant to one or more chemotherapeutic agents.
104. The method of claim 102 or 103, wherein the one or more chemotherapeutic or cytotoxic agents is dacarbazine, temozolomide, cisplatin, trosoxivas, fotemustine, IMCgp100, CTLA-4 inhibitors, PD-1 inhibitors, PD-L1 inhibitors, mitogen-activated protein kinase inhibitors, and/or protein kinase C inhibitors.
105. The method of any one of claims 87-104, wherein the melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematological cancer is melanoma.
106. The method of claim 105, wherein the melanoma is ocular pigment layer melanoma.
107. The method of claim 105, wherein the melanoma is mucosal melanoma.
108. The method of claim 105, wherein the melanoma is cutaneous melanoma.
109. The method of any one of claims 87-104, wherein the melanoma, prostate cancer, breast cancer, bone cancer, renal cell cancer, or hematologic cancer is hematologic cancer.
110. The method of claim 109, wherein the hematological cancer is multiple myeloma, large cell lymphoma, acute T-cell leukemia, acute myelogenous leukemia, myelodysplastic syndrome, immunoglobulin a lambda myeloma, diffuse mixed tissue cell and lymphocyte lymphoma, B-cell lymphoma, acute lymphoblastic leukemia, diffuse large cell lymphoma, or non-hodgkin's lymphoma.
111. The method of any one of claims 87-104, wherein the melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematological cancer is prostate cancer.
112. The method of any one of claims 87-104, wherein the melanoma, prostate cancer, breast cancer, bone cancer, renal cell cancer, or hematological cancer is breast cancer.
113. The method of claim 112, wherein the breast cancer is ER positive breast cancer, ER negative breast cancer, triple positive breast cancer, or triple negative breast cancer.
114. The method of any one of claims 87-104, wherein the melanoma, prostate cancer, breast cancer, bone cancer, renal cell cancer, or hematological cancer is bone cancer.
115. The method of claim 114, wherein the bone cancer is ewing's sarcoma.
116. The method of any one of claims 87-104, wherein the melanoma, prostate cancer, breast cancer, bone cancer, renal cell carcinoma, or hematological cancer is renal cell carcinoma.
117. The method of claim 116, wherein the renal cell carcinoma is a microphthalmia transcription factor (MITF) family translocation renal cell carcinoma.
CN202180089830.4A 2020-11-10 2021-11-10 Compounds and uses thereof Pending CN116745288A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US202063112126P 2020-11-10 2020-11-10
US63/112,126 2020-11-10
PCT/US2021/058865 WO2022103899A1 (en) 2020-11-10 2021-11-10 Compounds and uses thereof

Publications (1)

Publication Number Publication Date
CN116745288A true CN116745288A (en) 2023-09-12

Family

ID=81601667

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180089830.4A Pending CN116745288A (en) 2020-11-10 2021-11-10 Compounds and uses thereof

Country Status (10)

Country Link
US (1) US20230416248A1 (en)
EP (1) EP4244213A1 (en)
JP (1) JP2023551385A (en)
KR (1) KR20230106648A (en)
CN (1) CN116745288A (en)
AU (1) AU2021378949A1 (en)
CA (1) CA3198317A1 (en)
IL (1) IL302698A (en)
MX (1) MX2023005436A (en)
WO (1) WO2022103899A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023220219A1 (en) * 2022-05-11 2023-11-16 Foghorn Therapeutics Inc. Compounds and uses thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101402474B1 (en) * 2006-04-26 2014-06-19 제넨테크, 인크. Phosphoinositide 3-kinase inhibitor compounds and pharmaceutical compositions containing them
US9050345B2 (en) * 2013-03-11 2015-06-09 Bristol-Myers Squibb Company Pyrrolotriazines as potassium ion channel inhibitors
US9663502B2 (en) * 2013-12-20 2017-05-30 Lycera Corporation 2-Acylamidomethyl and sulfonylamidomethyl benzoxazine carbamates for inhibition of RORgamma activity and the treatment of disease
DK3174868T3 (en) * 2014-08-01 2021-11-08 Nuevolution As COMPOUNDS ACTIVE AGAINST BROMODOMAINS

Also Published As

Publication number Publication date
US20230416248A1 (en) 2023-12-28
IL302698A (en) 2023-07-01
MX2023005436A (en) 2023-07-25
JP2023551385A (en) 2023-12-08
CA3198317A1 (en) 2022-05-19
WO2022103899A1 (en) 2022-05-19
KR20230106648A (en) 2023-07-13
EP4244213A1 (en) 2023-09-20
AU2021378949A1 (en) 2023-06-15

Similar Documents

Publication Publication Date Title
EP3917934A2 (en) Compounds and uses thereof
US20220119378A1 (en) Compounds and uses thereof
US20230150974A1 (en) Compounds and uses thereof
US20230079819A1 (en) Compounds and uses thereof
EP4247815A1 (en) Compounds and uses thereof
US20230145003A1 (en) Compounds and uses thereof
EP4247381A1 (en) Compounds and uses thereof
KR20220133258A (en) Compounds and uses thereof
CN116745288A (en) Compounds and uses thereof
US20230121497A1 (en) Compounds and uses thereof
US20230365560A1 (en) Compounds and uses thereof
TW202411228A (en) Compounds and uses thereof
WO2023220134A1 (en) Pyrazine derivatives and uses thereof
JP2024516995A (en) Compounds and their uses
WO2023220137A1 (en) Pyrazine derivatives and uses thereof
WO2023220129A1 (en) Benzoyparazine pyrazines ane their uses
WO2022025880A1 (en) Compounds and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination